20.01.2015 Views

Chemistry and Biochemistry of HCA.pdf - AFBoard.com

Chemistry and Biochemistry of HCA.pdf - AFBoard.com

Chemistry and Biochemistry of HCA.pdf - AFBoard.com

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

10 J. Agric. Food Chem. 2002, 50, 10−22<br />

REVIEWS<br />

<strong>Chemistry</strong> <strong>and</strong> <strong>Biochemistry</strong> <strong>of</strong> (−)-Hydroxycitric Acid from<br />

Garcinia<br />

B. S. JENA, G.K.JAYAPRAKASHA, R.P.SINGH, AND K. K. SAKARIAH*<br />

Human Resource Development, Central Food Technological Research Institute, Mysore 570 013, India<br />

(-)-Hydroxycitric acid [(-)-<strong>HCA</strong>] is the principal acid <strong>of</strong> fruit rinds <strong>of</strong> Garcinia cambogia, Garcinia<br />

indica, <strong>and</strong> Garcinia atroviridis. (-)-<strong>HCA</strong> was shown to be a potent inhibitor <strong>of</strong> ATP citrate lyase (EC<br />

4.1.3.8), which catalyzes the extramitochondrial cleavage <strong>of</strong> citrate to oxaloacetate <strong>and</strong> acetyl-CoA:<br />

citrate + ATP + CoA f acetyl-CoA + ADP + P i + oxaloacetate. The inhibition <strong>of</strong> this reaction limits<br />

the availability <strong>of</strong> acetyl-CoA units required for fatty acid synthesis <strong>and</strong> lipogenesis during a lipogenic<br />

diet, that is, a diet high in carbohydrates. Extensive animal studies indicated that (-)-<strong>HCA</strong> suppresses<br />

the fatty acid synthesis, lipogenesis, food intake, <strong>and</strong> induced weight loss. In vitro studies revealed<br />

the inhibitions <strong>of</strong> fatty acid synthesis <strong>and</strong> lipogenesis from various precursors. However, a few clinical<br />

studies have shown controversial findings. This review explores the literature on a number <strong>of</strong> topics:<br />

the source <strong>of</strong> (-)-<strong>HCA</strong>; the discovery <strong>of</strong> (-)-<strong>HCA</strong>; the isolation, stereochemistry, properties, methods<br />

<strong>of</strong> estimation, <strong>and</strong> derivatives <strong>of</strong> (-)-<strong>HCA</strong>; <strong>and</strong> its biochemistry, which includes inhibition <strong>of</strong> the citrate<br />

cleavage enzyme, effects on fatty acid synthesis <strong>and</strong> lipogenesis, effects on ketogenesis, other<br />

biological effects, possible modes <strong>of</strong> action on the reduction <strong>of</strong> food intake, promotion <strong>of</strong> glycogenesis,<br />

gluconeogenesis, <strong>and</strong> lipid oxidation, (-)-<strong>HCA</strong> as weight-controlling agent, <strong>and</strong> some possible<br />

concerns about (-)-<strong>HCA</strong>, which provides a coherent presentation <strong>of</strong> scattered literature on (-)-<strong>HCA</strong><br />

<strong>and</strong> its plausible mechanism <strong>of</strong> action <strong>and</strong> is provocative <strong>of</strong> further research.<br />

Keywords: Garcinia cambogia; Garcinia indica; Garcinia atroviridis; (-)-hydroxycitric acid; ATP:citrate<br />

lyase; fatty acid synthesis; lipogenesis; appetite; antiobesity<br />

INTRODUCTION<br />

Garcinia (family: Guttiferae) is a large genus <strong>of</strong> polygamous<br />

trees or shrubs, distributed in tropical Asia, Africa, <strong>and</strong><br />

Polynesia. It consists <strong>of</strong> 180 species, <strong>of</strong> which ∼30 species are<br />

found in India (1). For the past several years, small <strong>and</strong> <strong>com</strong>plex<br />

molecules have been isolated from the various species <strong>of</strong><br />

Garcinia, which include xanthones <strong>and</strong> xanthone derivatives<br />

(2-4). However, the isolation <strong>of</strong> (-)-hydroxycitric acid [(-)-<br />

<strong>HCA</strong>] from a few species <strong>of</strong> Garcinia <strong>and</strong> its biological<br />

properties have attracted the attention <strong>of</strong> biochemists <strong>and</strong> health<br />

practitioners.<br />

The physiological <strong>and</strong> biochemical effects <strong>of</strong> (-)-<strong>HCA</strong> have<br />

been studied extensively for its unique regulatory effect on fatty<br />

acid synthesis, lipogenesis, appetite, <strong>and</strong> weight loss. The<br />

derivatives <strong>of</strong> (-)-<strong>HCA</strong> have been incorporated into a wide<br />

range <strong>of</strong> pharmaceutical preparations in <strong>com</strong>bination with other<br />

ingredients for the claimed purpose <strong>of</strong> enhancing weight loss,<br />

cardioprotection, correcting conditions <strong>of</strong> lipid abnormalities,<br />

<strong>and</strong> endurance in exercise (5-22).<br />

In the present review attempts have been made to pool,<br />

analyze, <strong>and</strong> summarize the available information on the<br />

chemical, physiological, <strong>and</strong> biochemical aspects <strong>of</strong> (-)-<strong>HCA</strong>.<br />

SOURCES OF (-)-<strong>HCA</strong><br />

(-)-<strong>HCA</strong> is found in the fruit rinds <strong>of</strong> certain species <strong>of</strong><br />

Garcinia, which include G. cambogia, G. indica, <strong>and</strong> G.<br />

atroViridis (23-25). These species thrive prolifically on the<br />

Indian subcontinent <strong>and</strong> in western Sri Lanka (1, 26).<br />

G. cambogia is a small or medium-sized tree with a rounded<br />

crown <strong>and</strong> horizontal or drooping branches; its leaves are dark<br />

green <strong>and</strong> shiny, elliptic obovate, 2-5 in. long <strong>and</strong> 1-3 in.<br />

broad; its fruits are ovoid, 2 in. in diameter, yellow or red when<br />

ripe with six to eight grooves, <strong>and</strong> the fruits have six to eight<br />

seeds surrounded by a succulent aril. The tree is found<br />

<strong>com</strong>monly in the evergreen forests <strong>of</strong> Western Ghats, from<br />

Konkan southward to Travancore, <strong>and</strong> in the Shola forests <strong>of</strong><br />

Nilgiris up to an altitude <strong>of</strong> 6000 ft. It flowers during the hot<br />

season, <strong>and</strong> fruits ripen during the rainy season. The seeds <strong>of</strong><br />

G. cambogia contain 31% edible fat.<br />

10.1021/jf010753k CCC: $22.00 © 2002 American Chemical Society<br />

Published on Web 11/29/2001


Reviews J. Agric. Food Chem., Vol. 50, No. 1, 2002 11<br />

G. indica is a slender evergreen tree with drooping branches;<br />

its leaves are ovate or oblong lanceolate, 2.5-3.5 in. long <strong>and</strong><br />

1-1.5 in. broad, dark green above <strong>and</strong> pale beneath; its fruits<br />

are globose or spherical, 1-1.5 in. diameter, dark purple when<br />

ripe <strong>and</strong> enclosing five to eight large seeds. The tree is found<br />

in the tropical rain forests <strong>of</strong> Western Ghats, from Konkan<br />

southward to Mysore, Coorg, <strong>and</strong> Wynaad. It flowers in<br />

November-February, <strong>and</strong> fruits ripen in April-May. The root<br />

is astringent. The seeds <strong>of</strong> the fruit have edible fat, <strong>com</strong>mercially<br />

known as Kokam butter<br />

G. atroViridis is a moderate-sized graceful tree, 30-50 ft high,<br />

found in the northeastern districts <strong>of</strong> upper Assam. Leaves are<br />

6-9 in. long <strong>and</strong> 2-3 in. broad, thickly coriaceous, glabrous,<br />

abruptly acuminate, <strong>and</strong> base contracted; its flowers are terminal,<br />

the female flower being solitary <strong>and</strong> large. The fruits are orange<br />

yellow, subglobose (3-4 in. across), fluted with a firmly<br />

textured outer rind <strong>and</strong> a rather thin <strong>and</strong> translucent pulp<br />

surrounding the seeds (1).<br />

The fruits <strong>of</strong> G. cambogia are too acidic to be eaten raw. A<br />

decoction <strong>of</strong> the fruit rind is given for rheumatism <strong>and</strong> bowel<br />

<strong>com</strong>plaints. It is also employed in veterinary medicine as a rinse<br />

for the diseases <strong>of</strong> the mouth in cattle. However, the fruit <strong>of</strong> G.<br />

indica has an agreeable flavor <strong>and</strong> a sweetish acid taste. It is<br />

used as a garnish to give an acid flavor to curries <strong>and</strong> also for<br />

preparing syrups during hot months. The fruit is also anthelmintic<br />

<strong>and</strong> useful in piles, dysentery, tumor, pains, <strong>and</strong> heart<br />

<strong>com</strong>plaints. Syrup from the fruit juice is given in bilious<br />

affections. The fruits <strong>of</strong> both species are valued for their dried<br />

rinds, which are used as a condiment for flavoring curries in<br />

place <strong>of</strong> tamarind or lemon. In Ceylon the dried fruit rinds <strong>of</strong><br />

G. cambogia are used along with salt in the curing <strong>of</strong> fish (1).<br />

The fruit rinds <strong>of</strong> G. atroViridis are also too acidic to be eaten<br />

raw, but the taste is excellent when stewed with sugar. In Malay,<br />

the sun-dried rinds <strong>of</strong> under-ripe fruits are sold as a sour relish<br />

for use in curries in place <strong>of</strong> tamarind <strong>and</strong> for the dressing <strong>of</strong><br />

fish. The fruit is also used as a fixative with alum in the dyeing<br />

<strong>of</strong> silk. A decoction from leaves <strong>and</strong> roots is used in the<br />

treatment <strong>of</strong> earache (1).<br />

CHEMISTRY OF (-)-<strong>HCA</strong><br />

Discovery <strong>of</strong> ())-<strong>HCA</strong>. The dried rind <strong>of</strong> the fruit <strong>of</strong> G.<br />

cambogia popularly known as “Malabar tamarind” is extensively<br />

used all over the west coast <strong>of</strong> South India for culinary purposes<br />

<strong>and</strong> <strong>com</strong>mercially for “Colombo curing” <strong>of</strong> fish (23, 27, 28).<br />

The organic acids present in the fruit have been held responsible<br />

for the bacteriostatic effect <strong>of</strong> the pickling medium by lowering<br />

the pH (27). The fruit contains ∼30% acid calculated as citric<br />

acid on a dry weight basis (28). The organic acids present have<br />

been mistakenly identified as tartaric acid <strong>and</strong> citric acids (27,<br />

29). The aqueous extracts <strong>of</strong> the fruit showed two predominant<br />

acid spots on paper chromatograms run on different solvents,<br />

which were very near to tartaric <strong>and</strong> citric acids, but there was<br />

always a significant small difference in the R f values in all<br />

solvent systems. Analysis <strong>of</strong> the fruit juice by use <strong>of</strong> an ion<br />

exchange column as described by Palmer (30) showed the largest<br />

peak to be the citric acid region, but the acids failed the cream<br />

<strong>of</strong> tartar test for tartaric acid <strong>and</strong> the pentabromoacetone test<br />

for citric acid (28).<br />

Lewis <strong>and</strong> Neelakantan (23) isolated the principal acid in the<br />

fruit rinds <strong>of</strong> G. cambogia <strong>and</strong> identified it as (-)-<strong>HCA</strong> on the<br />

basis <strong>of</strong> chemical <strong>and</strong> spectroscopic studies. Identification <strong>and</strong><br />

separation <strong>of</strong> the hydroxycitric acid on Whatman No. 1 paper<br />

were performed using n-butanol/acetic acid/water (4:1:5) <strong>and</strong><br />

n-propanol/formic acid/water (4:1:5). The spots were identified<br />

by spraying with 5% metavanadate. Upon saponification <strong>of</strong> the<br />

Figure 1. Structures <strong>of</strong> hydroxycitric acid isomers.<br />

Figure 2. Structures <strong>of</strong> hydroxycitric acid lactones.<br />

acid mixture with excess alkali <strong>and</strong> passing it through a column<br />

<strong>of</strong> ion exchange resin (Zeocarb 215), the eluate showed only<br />

one lower spot (R f ) 0.34) corresponding to the free (-)-<strong>HCA</strong>.<br />

On concentration the eluate gave only one upper spot (R f )<br />

0.46) corresponding to the lactone. Fruit extracts showed two<br />

predominant acid spots on chromatograms with two different<br />

solvent systems. On titration <strong>of</strong> this material with alkali, using<br />

phenolphthalein, two different end-points were obtained, in the<br />

cold <strong>and</strong> after boiling, showing the characteristics <strong>of</strong> lactones.<br />

The two spots on chromatograms were identified as hydroxycitric<br />

acid <strong>and</strong> its lactone (Figures 1 <strong>and</strong> 2). It was thus clear<br />

that the two spots on the chromatograms are those <strong>of</strong> γ-hydroxy<br />

acid <strong>and</strong> its lactone <strong>and</strong> not those <strong>of</strong> tartaric acid <strong>and</strong> citric acids.<br />

Isolation. Lewis <strong>and</strong> Neelakantan (23) isolated this (-)-<strong>HCA</strong><br />

on a large scale from the dried rinds <strong>of</strong> G. cambogia. The<br />

method consisted <strong>of</strong> extracting the acid by cooking the raw<br />

material with water under pressure (10 lb/in. 2 for 15 min). The<br />

extract was concentrated, <strong>and</strong> pectin was removed by alcohol<br />

precipitation. The clear filtrate was neutralized with alkali,<br />

passed through cation exchange resin for recovery <strong>of</strong> the acid,<br />

which was concentrated <strong>and</strong> dried. The crude dried mass was<br />

extracted with ether <strong>and</strong> recrystallized to give small needleshaped<br />

crystals <strong>of</strong> lactone. Lewis (24) reported another method<br />

for the isolation <strong>of</strong> (-)-<strong>HCA</strong> from G. cambogia using acetone.<br />

The acetone extract was concentrated <strong>and</strong> the acid taken up in<br />

water. The aqueous solution yielded the lactone on evaporation.<br />

M<strong>of</strong>fett et al. (31) have developed a process for the aqueous<br />

extraction <strong>of</strong> (-)-<strong>HCA</strong> from Garcinia rinds. The extract was<br />

loaded onto an anion exchange column for adsorption <strong>of</strong> (-)-<br />

<strong>HCA</strong>, <strong>and</strong> it was eluted with sodium/potassium hydroxide for<br />

release <strong>of</strong> (-)-<strong>HCA</strong>. The extract was passed through a cation<br />

exchange column to yield a free acid. Guthrie <strong>and</strong> Kierstead<br />

(9, 10) <strong>and</strong> M<strong>of</strong>fett et al. (32) have reported the preparation <strong>of</strong><br />

(-)-<strong>HCA</strong> concentrate from Garcinia rinds with 23-54% (-)-<br />

<strong>HCA</strong> <strong>and</strong> 6-20% lactone.


12 J. Agric. Food Chem., Vol. 50, No. 1, 2002 Reviews<br />

Table 1. Comparison <strong>of</strong> Physical Properties <strong>of</strong> <strong>HCA</strong> <strong>and</strong> Lactones<br />

from Garcinia <strong>and</strong> Hibiscus (23)<br />

property<br />

free<br />

acid<br />

Garcinia<br />

lactone<br />

free<br />

acid<br />

Hibiscus<br />

lactone<br />

mp (°C) 178 183<br />

[R] 20 D (deg) −20 100 122 31<br />

crystal shape needles needles<br />

hygroscopicity slight high<br />

solubility<br />

high in alcohol<br />

<strong>and</strong> water; fair<br />

in ether<br />

paper chromatogr (R f)<br />

butanol/formic acid/H 2O 0.24 0.42 0.15 0.39<br />

propanol/acetic acid/H 2O 0.26 0.36 0.35 0.26<br />

metav<strong>and</strong>ate spray (5%) yellow reddish orange yellow yellow<br />

high in water<br />

<strong>and</strong> alcohol;<br />

slight in ether<br />

Stereochemistry. Hydroxycitric acid (1,2-dihydroxypropane-<br />

1,2,3-tricarboxylic acid) has two asymmetric centers; hence, two<br />

pairs <strong>of</strong> diastereoisomers or four different isomers are possible<br />

(Figure 1). Martius <strong>and</strong> Maue (33) have synthesized the four<br />

possible stereoisomers <strong>of</strong> hydroxycitrate. One <strong>of</strong> these isomers<br />

occurs in Garcinia (Figure 1, I) <strong>and</strong> another in Hibiscus species<br />

(Figure 1, II) (24). The absolute configurations <strong>of</strong> the hydroxycitric<br />

acid lactones, hibiscus acid <strong>and</strong> garcinia acid, were<br />

determined to be (2S,3R)- <strong>and</strong> (2S,3S)-2-hydroxycitric acid-2,5-<br />

lactone, respectively (Figure 2). The absolute configuration is<br />

determined from Hudson’s lactone rule, optical rotatory dispersion<br />

curves, circular dichrosim curves, <strong>and</strong> calculation <strong>of</strong> partial<br />

molar rotations (34). Glusker et al. (35, 36) have reported the<br />

structure <strong>and</strong> absolute configuration <strong>of</strong> the calcium hydroxycitrate<br />

<strong>and</strong> (-)-<strong>HCA</strong> lactone by X-ray crystallography. Stallings<br />

et al. (37) have reported the crystal structures <strong>of</strong> the ethylenediamine<br />

salts <strong>of</strong> diastereoisomeric hydroxycitrates.<br />

Properties <strong>of</strong> ())-<strong>HCA</strong> <strong>and</strong> Lactone. The physical properties<br />

<strong>of</strong> (-)-<strong>HCA</strong> <strong>and</strong> lactones (Figure 2) from Garcinia <strong>and</strong><br />

Hibiscus are presented in Table 1. The equivalent weight <strong>of</strong><br />

pure lactone is 69, as determined by alkali titration or silver<br />

salt de<strong>com</strong>position. The IR spectra <strong>of</strong> the ethyl ester showed<br />

ester <strong>and</strong> hydroxyl groups at 5.41-5.76 <strong>and</strong> 2.74-2.79 µ,<br />

respectively (23). The structure <strong>of</strong> the (-)-<strong>HCA</strong> lactone was<br />

further established by IR <strong>and</strong> 1 H NMR spectroscopy. The (-)-<br />

<strong>HCA</strong> lactone displayed strong IR b<strong>and</strong>s at 3200, 1760, <strong>and</strong> 1680<br />

cm -1 . 1 H NMR spectra <strong>of</strong> the (-)-<strong>HCA</strong> lactone showed two<br />

protons at the γ-carbon, which give an AB quartet at δ 2.53<br />

<strong>and</strong> δ 2.74 with J ) 17.1 Hz, <strong>and</strong> one proton at the R-carbon<br />

showing a singlet at δ 5.15 (38).<br />

Estimation <strong>of</strong> ())-<strong>HCA</strong>. Free (-)-<strong>HCA</strong> leads to the<br />

formation <strong>of</strong> (-)-<strong>HCA</strong> lactone during concentration <strong>and</strong> evaporation.<br />

The presence <strong>of</strong> minor organic acids such as citric acid,<br />

tartaric acid, <strong>and</strong> malic acid in the Garcinia fruits <strong>and</strong> the lack<br />

<strong>of</strong> <strong>of</strong>ficial methods for the assay <strong>of</strong> (-)-<strong>HCA</strong> have created<br />

confusion <strong>and</strong> disagreement among analysts (39). The existing<br />

method for the determination <strong>of</strong> (-)-<strong>HCA</strong> content in G.<br />

cambogia extract involves acid-base titration, which gives the<br />

total acidity <strong>of</strong> the extract (40). However, in this method the<br />

concentrations <strong>of</strong> (-)-<strong>HCA</strong> <strong>and</strong> lactone cannot be estimated<br />

separately.<br />

Lowenstein <strong>and</strong> Brunengraber (25) have estimated the hydroxycitrate<br />

content <strong>of</strong> the fruit <strong>of</strong> G. cambogia by gas<br />

chromatography (GC). GC estimation involves the conversion<br />

<strong>of</strong> acid to volatile silyl derivative. They used an OV-17 GC<br />

column (3 m × 3 mm i.d.). The column was run at 145 °C<br />

using nitrogen as the carrier gas (40 mL/min) with an injection<br />

port temperature <strong>of</strong> 250 °C <strong>and</strong> a detector temperature <strong>of</strong> 300<br />

°C. (-)-<strong>HCA</strong> lactone is the major constituent <strong>of</strong> the extract,<br />

Figure 3. Structures <strong>of</strong> hydroxycitric acid derivatives.<br />

<strong>and</strong> the recrystallized <strong>com</strong>pound contains


Reviews J. Agric. Food Chem., Vol. 50, No. 1, 2002 13<br />

for the purified enzyme than the natural substrate, that is, citrate,<br />

<strong>and</strong> the K i <strong>of</strong> (-)-<strong>HCA</strong> for citrate cleavage enzyme was between<br />

0.2 <strong>and</strong> 0.6 µM depending on the conditions (47, 48). Later on,<br />

Cheema-Dhadli et al. (49) found inhibition <strong>of</strong> citrate cleavage<br />

enzyme by both free (-)-<strong>HCA</strong> (K i ) 8 µM) <strong>and</strong> (-)-<strong>HCA</strong><br />

lactone (K i ) 50-100 µM). The lactone <strong>of</strong> (-)-<strong>HCA</strong> was found<br />

to be a very less effective inhibitor <strong>of</strong> citrate cleavage enzyme.<br />

In rat brain synaptosomes, (-)-<strong>HCA</strong> was shown to be a potent<br />

inhibitor <strong>of</strong> citrate cleavage enzyme (50). Sullivan et al. (51)<br />

<strong>and</strong> Stallings et al. (37), in similar studies, observed that <strong>of</strong> four<br />

isomers <strong>of</strong> <strong>HCA</strong> (Figure 1), (-)-<strong>HCA</strong> was the only potent<br />

inhibitor <strong>of</strong> ATP:citrate lyase. (-)-<strong>HCA</strong> was found to inhibit<br />

partially purified ATP:citrate lyase from human liver <strong>com</strong>petitively<br />

with respect to citrate (K i ) 3 × 10 -4 M) (52).<br />

The discovery <strong>of</strong> the powerful inhibition <strong>of</strong> citrate cleavage<br />

enzyme by (-)-<strong>HCA</strong> provides a valuable tool for the study <strong>of</strong><br />

the metabolic role <strong>of</strong> citrate cleavage reaction (47).<br />

The biological effect <strong>of</strong> (-)-<strong>HCA</strong> stems from the inhibition<br />

<strong>of</strong> extramitochondrial cleavage <strong>of</strong> citrate to oxaloacetate <strong>and</strong><br />

acetyl-CoA. The ultimate source <strong>of</strong> all the carbon atoms <strong>of</strong> fatty<br />

acids is acetyl-CoA, formed in the mitochondria by the oxidative<br />

decarboxylation <strong>of</strong> pyruvate, the oxidative degradation <strong>of</strong> some<br />

<strong>of</strong> the amino acids, or the β-oxidation <strong>of</strong> long-chain fatty acids.<br />

The synthesis <strong>of</strong> fatty acids is maximal when carbohydrate is<br />

abundant <strong>and</strong> the level <strong>of</strong> fatty acids is low. The conversion <strong>of</strong><br />

carbohydrate into fat involves the oxidation <strong>of</strong> pyruvate to<br />

acetyl-CoA. Fatty acids are synthesized in the cytosol, whereas<br />

acetyl-CoA is formed from pyruvate in mitochondria. For fatty<br />

acid biosynthesis, the acetyl group <strong>of</strong> acetyl-CoA must be<br />

transferred from the mitochondria to the cytosol. Acetyl-CoA<br />

as such cannot pass out <strong>of</strong> the mitochondria into the cytosol.<br />

The barrier to acetyl-CoA is bypassed by citrate, which carries<br />

acetyl groups across the inner mitochondrial membrane. Citrate<br />

is formed in the mitochondrial matrix by the condensation <strong>of</strong><br />

acetyl-CoA with oxaloacetate. When present at high levels,<br />

citrate is transported to cytosol, via the tricarboxylate transport<br />

system. In cytosol, acetyl-CoA is regenerated from citrate by<br />

the ATP:citrate lyase, which catalyzes the following reaction:<br />

citrate + ATP + CoA f acetyl-CoA + ADP + P i +<br />

oxaloacetate (53-57). ATP:citrate lyase is widely distributed<br />

in animal tissues (58). ATP:citrate lyase has been suggested to<br />

play a physiological role in lipogenesis from carbohydrate (59)<br />

<strong>and</strong> gluconeogenesis (60). The changes in activity <strong>of</strong> citrate<br />

cleavage enzyme correlate with changes in the rate <strong>of</strong> fatty acid<br />

synthesis <strong>and</strong> provide evidence for the involvement <strong>of</strong> the citrate<br />

cleavage reaction in fatty acid synthesis (61, 62). The activity<br />

<strong>of</strong> citrate cleavage enzyme varies in accordance with the<br />

nutritional status <strong>of</strong> the animal. Thus, during starvation or when<br />

fed on a high-fat diet, the enzyme levels fall drastically <strong>and</strong> on<br />

feeding <strong>of</strong> a high-carbohydrate diet elevated levels <strong>of</strong> ATP:<br />

citrate lyase were detected (61, 63). Evidence also exists<br />

demonstrating that >80% <strong>of</strong> the extramitochondrial acetyl-CoA<br />

produced from pyruvate in rat liver mitochondria is supplied<br />

via ATP:citrate lyase (64).<br />

Effect <strong>of</strong> ())-<strong>HCA</strong> on Fatty Acid Synthesis <strong>and</strong> Lipogenesis.<br />

(-)-<strong>HCA</strong> being a potent inhibitor <strong>of</strong> ATP:citrate lyase,<br />

which catalyzes the extramitochondrial cleavage <strong>of</strong> citrate to<br />

oxaloacetate <strong>and</strong> acetyl-CoA, limits the availability <strong>of</strong> acetyl-<br />

CoA units required for fatty acid synthesis <strong>and</strong> lipogenesis<br />

(65-68). Many studies demonstrated both in vitro <strong>and</strong> in vivo<br />

that (-)-<strong>HCA</strong> suppresses the de novo fatty acid synthesis <strong>and</strong><br />

lipogenesis.<br />

In a cell-free system, consisting <strong>of</strong> particle-free cytoplasm<br />

<strong>and</strong> mitochondria prepared from rat liver, (-)-<strong>HCA</strong> was shown<br />

to inhibit fatty acid synthesis from citrate <strong>and</strong> also from [ 14 C]-<br />

alanine by measuring the incorporation <strong>of</strong> 3 H 2 O <strong>and</strong> 14 C(53).<br />

Sullivan et al. (65) studied the effect <strong>of</strong> stereoisomers <strong>of</strong><br />

hydroxycitrate (Figure 1) on the rate <strong>of</strong> lipogenesis in rat liver.<br />

They found that only (-)-<strong>HCA</strong> significantly decreased the<br />

conversion <strong>of</strong> [ 14 C]citrate into lipid in the liver high-speed<br />

supernatant <strong>and</strong> the conversion <strong>of</strong> [ 14 C]alanine into lipid in vivo.<br />

Beynen <strong>and</strong> Geelen (69) observed the inhibition <strong>of</strong> fatty acid<br />

synthesis from glucose by (-)-<strong>HCA</strong> in isolated hepatocytes,<br />

but it did not affect fatty acid synthesis from acetate; rather,<br />

(-)-<strong>HCA</strong> was shown to stimulate fatty acid synthesis from<br />

acetate (70). This effect was observed both in the isolated<br />

perfused rat liver <strong>and</strong> in the homogenates <strong>of</strong> this organ <strong>and</strong> was<br />

assumed to be due to an activation <strong>of</strong> acetyl-CoA carboxylase<br />

in analogy to other tricarboxylates (71, 72). Moreover, the<br />

findings <strong>of</strong> Hackenschmidt et al. (73) showed that (-)-<strong>HCA</strong> is<br />

a positive effector <strong>of</strong> acetyl-CoA carboxylase <strong>and</strong> similarity <strong>of</strong><br />

(-)-<strong>HCA</strong> <strong>and</strong> citrate activation <strong>of</strong> enzyme activity was apparent<br />

in the time for maximal stimulation. It was concluded that (-)-<br />

<strong>HCA</strong> acts as an inhibitor <strong>of</strong> lipogenesis only if cytoplasmic<br />

acetyl-CoA is produced by citrate cleavage enzyme reaction,<br />

but it will activate fatty acid synthesis wherever an alternate<br />

source <strong>of</strong> acetyl-CoA, for example, acetate, is available.<br />

The increase in lipogenesis in brown adipose in response to<br />

glucose feeding was inhibited by (-)-<strong>HCA</strong> (74). In starved rats,<br />

(-)-<strong>HCA</strong> did not inhibit the basal rates <strong>of</strong> brown fat lipogenesis<br />

<strong>and</strong> glucose utilization in response to insulin. As basal rates <strong>of</strong><br />

lipogenesis were not inhibited by (-)-<strong>HCA</strong>, it is suggested that<br />

acetate may be a lipogenic substrate for brown fat in starvation<br />

(75). Smith (76) reported that rabbit adipose tissue has a low<br />

capacity for de novo fatty acid synthesis from glucose but a<br />

high capacity for synthesis from pyruvate <strong>and</strong> acetate. Experiments<br />

with (-)-<strong>HCA</strong> have demonstrated that citrate cleavage<br />

enzyme is an obligatory enzyme in lipogenesis from pyruvate<br />

<strong>and</strong> that the lipogenic system <strong>of</strong> rabbit adipose tissue resembles<br />

that <strong>of</strong> a ruminant in that it is adapted to utilize acetate rather<br />

than glucose.<br />

(-)-<strong>HCA</strong> partially inhibited the incorporation <strong>of</strong> 3 H from<br />

3 H 2 O <strong>and</strong> 14 C from [U- 14 C]lactate into saponifiable fatty acid<br />

in the hepatocytes from fed rats, but incorporation <strong>of</strong> 14 C from<br />

[ 14 C]proline was <strong>com</strong>pletely inhibited. However, similar <strong>com</strong>plete<br />

inhibition <strong>of</strong> incorporation <strong>of</strong> 14 C from [ 14 C]proline was<br />

observed in fed rats, glycogen-depleted hepatocytes, or hepatocytes<br />

from starved rats (77). In another observation, (-)-<strong>HCA</strong><br />

depressed lipogenesis in hepatocytes from fed rats incubated<br />

with lactate plus pyruvate by ∼51% but had little effect on<br />

lipogenesis in glycogen-depleted hepatocytes similarly incubated.<br />

There was a parallel inhibition <strong>of</strong> incorporation <strong>of</strong> 14 C<br />

from [U- 14 C]lactate to fatty acids, <strong>and</strong> lipogenesis was measured<br />

with 3 H 2 O in each case. Thus, depletion <strong>of</strong> glycogen or<br />

conceivably the process <strong>of</strong> glycogen depletion causes a change<br />

in the rate-limiting step(s) for lipogenesis from lactate (78).<br />

(-)-Hydroxycitrate markedly reduced tritiated water incorporation<br />

into fatty acid by lung tissue slices (79). Starving rats<br />

for 72 h markedly reduced in vivo 3 H 2 O incorporation into<br />

pulmonary lipids. Intraperitoneal injection <strong>of</strong> (-)-<strong>HCA</strong> prior<br />

to 3 H 2 O administration also markedly reduced isotope incorporation<br />

into pulmonary lipid fractions <strong>of</strong> fed rats. It has been<br />

concluded that in vivo synthesis <strong>of</strong> fatty acids was affected by<br />

the nutritional state <strong>of</strong> the animal <strong>and</strong> citrate appears to be a<br />

significant source <strong>of</strong> cytoplasmic acetyl-CoA for the de novo<br />

pulmonary lipogenesis in the fed rats (80). Sheehan <strong>and</strong> Yeh<br />

(81) observed that (-)-<strong>HCA</strong> inhibited fatty acid synthesis in<br />

neonatal rat lung from glucose, pyruvate, <strong>and</strong> β-hydroxybutyrate


14 J. Agric. Food Chem., Vol. 50, No. 1, 2002 Reviews<br />

by 88, 70, <strong>and</strong> 60%, respectively, but had no effect on that from<br />

acetoacetate. Lipogenesis from β-hydroxybutyrate involves both<br />

the cytoplasmic <strong>and</strong> citrate pathways. Crabtree et al. (82)<br />

observed that the fluxes <strong>of</strong> butyrate to acetate <strong>and</strong> fatty acids<br />

in rat hepatocytes were inhibited to a similar extent by (-)-<br />

<strong>HCA</strong> with no significant effect on butyrate uptake. Because<br />

butyrate is activated only in the mitochondria <strong>of</strong> rat liver (83)<br />

<strong>and</strong> (-)-<strong>HCA</strong> did not significantly inhibit butyrate uptake, this<br />

parallel inhibition <strong>of</strong> the flux <strong>of</strong> butyrate to acetate <strong>and</strong> fatty<br />

acids strongly suggests that the production <strong>of</strong> acetate occurs in<br />

the cytoplasm. (-)-<strong>HCA</strong> could not be expected to inhibit the<br />

production <strong>of</strong> acetate via the mitochondrial hydrolase. Experiments<br />

with (-)-<strong>HCA</strong> indicate that the major route for conversion<br />

<strong>of</strong> leucine carbon to lipid in rat mammary gl<strong>and</strong> acini is via<br />

citrate translocation from the mitochondria (84). Mathias et al.<br />

(85) reported that in hepatocytes isolated from female rats, mealfed<br />

a high-glucose diet, (-)-<strong>HCA</strong> depressed the incorporation<br />

<strong>of</strong> 3 H 2 O, [ 14 C]alanine, [2- 14 C]leucine into fatty acids <strong>and</strong><br />

cholesterol.<br />

Hood et al. (86) have shown that (-)-<strong>HCA</strong> reduced the<br />

synthesis <strong>of</strong> fatty acids from lactate <strong>and</strong> glucose in bovine<br />

adipose tissue <strong>and</strong> rat adipose tissue, respectively, <strong>and</strong> suggested<br />

that the conversion <strong>of</strong> lactate to fatty acids probably occurs by<br />

way <strong>of</strong> citrate. In the (-)-<strong>HCA</strong>-treated rat liver cells, there was<br />

a decrease in the formation <strong>of</strong> phospholipds <strong>and</strong> triglycerides<br />

from lactate (87). Vicario <strong>and</strong> Medina (88) have also reported<br />

the inhibition <strong>of</strong> lipogenesis from lactate in rat brain by (-)-<br />

<strong>HCA</strong>, <strong>and</strong> the transfer <strong>of</strong> lactate carbons through the mitochondrial<br />

membrane is ac<strong>com</strong>panied by the translocation <strong>of</strong> citrate.<br />

(-)-<strong>HCA</strong> inhibited lipogenesis in rat brain slices (89, 90)<br />

<strong>and</strong> in freshly isolated mouse preputial gl<strong>and</strong> (91). (-)-<strong>HCA</strong><br />

equivalently reduced the biosynthesis <strong>of</strong> triglycerides, phospholipids,<br />

cholesterol, diglycerides, cholesteryl esters, <strong>and</strong> free<br />

fatty acids in isolated liver cells from normal <strong>and</strong> hyperlipidemic<br />

rats (92). (-)-<strong>HCA</strong> decreased the rate <strong>of</strong> cholesterol production<br />

in hepatocytes from fed rats by interfering with the flow <strong>of</strong><br />

substrate into the sterol biosynthetic pathway (93). Measurement<br />

<strong>of</strong> the weight <strong>of</strong> desmosterol produced during its biosynthesis<br />

in the presence <strong>of</strong> tritiated water <strong>and</strong> triparanol has permitted a<br />

direct determination <strong>of</strong> the relative flux <strong>of</strong> carbon <strong>and</strong> tritium<br />

(the H/C ratio) into sterol in hepatocytes. The H/C ratio, which<br />

increased with the time <strong>of</strong> incubation, was shown to decrease<br />

with (-)-<strong>HCA</strong>. The depression <strong>of</strong> biosynthesis <strong>of</strong> long-chain<br />

fatty acids, 3-β-hydroxysterol, <strong>and</strong> cholesterol by (-)-<strong>HCA</strong> was<br />

confirmed in a perfused rat liver system (70, 94, 95). The<br />

inhibition <strong>of</strong> fatty acid synthesis by (-)-<strong>HCA</strong> was associated<br />

with the decreased rate <strong>of</strong> choline incorporation into desaturated<br />

phosphatidylcholine in fetal rat lung (96). Berkhout et al. (97)<br />

tested (-)-<strong>HCA</strong> in Hep G2 cells for effects on cholesterol<br />

homeostasis. After 2.5 <strong>and</strong> 18 h <strong>of</strong> incubation with (-)-<strong>HCA</strong><br />

at a concentration <strong>of</strong> g0.5 mM, incorporation <strong>of</strong> [1,5- 14 C]citrate<br />

into fatty acids <strong>and</strong> cholesterol was strongly inhibited, which<br />

may reflect an effective inhibition <strong>of</strong> ATP:citrate lyase. Preincubation<br />

with a higher concentration <strong>of</strong> (-)-<strong>HCA</strong> increased the<br />

cellular low-density lipoprotein (LDL) receptor activity as<br />

determined by the receptor-mediated association <strong>and</strong> degradation.<br />

Measurements <strong>of</strong> receptor-mediated binding versus LDL<br />

concentration suggested that this increase might be due to an<br />

increase in the number <strong>of</strong> LDL receptors. A simultaneous<br />

increase <strong>of</strong> the enzyme levels <strong>of</strong> 3-hydroxy-3-methylglutaryl-<br />

CoA (HMG) reductase was also observed in the same condition.<br />

These results suggest that the increase in HMG-CoA reductase<br />

<strong>and</strong> LDL receptor are initiated by the decreased flux <strong>of</strong> carbon<br />

units in cholesterol synthetic pathway, owing to inhibition <strong>of</strong><br />

ATP citrate lyase.<br />

Hildebr<strong>and</strong>t et al. (98) studied the utilization <strong>and</strong> preferred<br />

metabolic pathway <strong>of</strong> ketone bodies for the synthesis <strong>of</strong> lipids<br />

by freshly isolated Morris Hepatoma 777 cells, which are known<br />

to actively convert ketone bodies to cholesterol <strong>and</strong> fatty acid.<br />

On the basis <strong>of</strong> the results with (-)-<strong>HCA</strong>, these authors found<br />

that the metabolic pathway for acetoacetate conversion to lipids<br />

is exclusively cytoplasmic, whereas that for 3-hydroxybutyrate<br />

involves both extra- <strong>and</strong> intramitochondrial <strong>com</strong>partments.<br />

Acetyl-L-carnitine is known as a modulator <strong>of</strong> metabolic<br />

function. Lligona-Trulla et al. (99) have studied lipogenesis in<br />

different cell lines <strong>and</strong> in rat hepatocytes <strong>and</strong> observed that the<br />

flux <strong>of</strong> acetyl-L-carnitine (ALC) to lipid was increased, not<br />

decreased, by (-)-<strong>HCA</strong>. In contrast, this inhibitor dramatically<br />

decreased the flux <strong>of</strong> glucose to lipid. These results indicate<br />

that the flux <strong>of</strong> acetyl-L-carnitine to lipid can bypass citrate <strong>and</strong><br />

utilize cytosolic acetyl-CoA synthesis, <strong>and</strong> it was shown that<br />

when (-)-<strong>HCA</strong> was used, the capacity <strong>of</strong> ALC to supply acetyl<br />

units for de novo lipogenesis could be increased significantly.<br />

Because ALC provides acetyl units for metabolic activities<br />

without the cost <strong>of</strong> ATP hydrolysis, this increased capacity may<br />

also represent a mechanism by which ALC can provide acetyl<br />

units in metabolically <strong>com</strong>promised situations. In insulindifferentiated<br />

3T3L1 cells, (-)-<strong>HCA</strong> was shown to inhibit<br />

lipogenesis <strong>and</strong> stimulate lipolysis (100).<br />

Incorporation <strong>of</strong> 3 H from 3 H 2 O was used to measure the rate<br />

<strong>of</strong> fatty acid synthesis in rat liver. In vivo inhibition <strong>of</strong> the rate<br />

<strong>of</strong> fatty acid synthesis in the rat liver was demonstrated after<br />

intraperitoneal/intravenous administration <strong>of</strong> (-)-<strong>HCA</strong> (101).<br />

The in vivo rate <strong>of</strong> lipogenesis was markedly decreased from<br />

[ 14 C]alanine following the administration <strong>of</strong> (-)-<strong>HCA</strong> either<br />

intraperitoneally or intravenously. Fatty acid <strong>and</strong> cholesterol<br />

syntheses were significantly inhibited by the oral administration<br />

<strong>of</strong> (-)-<strong>HCA</strong> only when the <strong>com</strong>pound was given before the<br />

feeding period (65). The oral administration <strong>of</strong> (-)-<strong>HCA</strong> to rats<br />

significantly depressed the in vivo lipogenic rates in a dosedependent<br />

manner in the liver, adipose tissue, <strong>and</strong> small intestine<br />

<strong>and</strong> caused significant reductions in body-weight gain, food<br />

consumption, <strong>and</strong> total body lipid (66, 67). (-)-Hydroxycitric<br />

acid <strong>and</strong> (+)-allo-hydroxycitric acid were also investigated (102)<br />

for their effects on in vivo lipid synthesis under conditions <strong>of</strong><br />

either high-carbohydrate feeding or 24 h fasting, <strong>and</strong> it was<br />

found that in fed rat, (-)-<strong>HCA</strong> significantly reduced the<br />

incorporation <strong>of</strong> labeled H 2 O <strong>and</strong> alanine into fatty acids <strong>and</strong><br />

cholesterol. An increased rate <strong>of</strong> incorporation <strong>of</strong> labeled H 2 O<br />

into fatty acids but no change in cholesterol synthesis in the<br />

fasted rate suggested that (-)-<strong>HCA</strong> might be an activator <strong>of</strong><br />

acetyl-CoA carboxylase. (+)-allo-Hydroxycitric acid was ineffective<br />

in modulating the rates <strong>of</strong> fatty acid synthesis under either<br />

nutritional condition. Both (-)-hydroxycitric acid <strong>and</strong> (+)-allohydroxycitric<br />

acid were shown to be in vitro activators <strong>of</strong> acetyl-<br />

CoA carboxylase, the former being a stronger activator. Thus,<br />

stereospecificity <strong>of</strong> the hydroxycitrate isomers was demonstrated<br />

in both the inhibition <strong>and</strong> stimulation <strong>of</strong> fatty acid synthesis<br />

possibly occurring at the level <strong>of</strong> acetyl-CoA carboxylase.<br />

Because the increase <strong>of</strong> citrate level could have allosterically<br />

activated acetyl-CoA carboxylase (103), this activation <strong>of</strong> acetyl-<br />

CoA carboxylase may be due to the increased level <strong>of</strong> citrate<br />

caused by the inhibition <strong>of</strong> citrate cleavage enzyme with (-)-<br />

<strong>HCA</strong>.<br />

In mature rat, the gold thioglucose-induced obese mouse, <strong>and</strong><br />

the ventromedial hypothalamic lesioned obese rat, food intake,<br />

body-weight gain, <strong>and</strong> depression <strong>of</strong> body lipid levels were


Reviews J. Agric. Food Chem., Vol. 50, No. 1, 2002 15<br />

reduced significantly by the chronic administration <strong>of</strong> (-)-<strong>HCA</strong><br />

(104). Chee et al. (105) have observed species-specific response<br />

to (-)-<strong>HCA</strong>. Acute administration <strong>of</strong> (-)-<strong>HCA</strong> caused the<br />

depression <strong>of</strong> in vivo rates <strong>of</strong> fatty acid synthesis in the livers<br />

<strong>of</strong> chickens <strong>and</strong> rats. Chickens appeared to be more sensitive<br />

to the inhibitory effects <strong>of</strong> (-)-<strong>HCA</strong> than did the rats. Oral<br />

administration <strong>of</strong> (-)-<strong>HCA</strong> significantly reduced the rate <strong>of</strong> in<br />

vivo hepatic fatty acids <strong>and</strong> cholesterol synthesis, <strong>and</strong> there was<br />

a reduction in serum triglyceride <strong>and</strong> cholesterol levels in<br />

normolipidemic rats. It was also observed that (-)-<strong>HCA</strong><br />

significantly reduced the hypertriglyceridemia <strong>and</strong> hyperlipogenesis<br />

in genetically obese Zucker rats, fructose-treated rats<br />

with elevated triglyceride, <strong>and</strong> Triton-treated rats (92, 106).<br />

Young Zucker lean (Fa/-) <strong>and</strong> obese (fa/fa) female rats were<br />

fed with (-)-<strong>HCA</strong>, <strong>and</strong> decreases in body weight, food intake,<br />

percent <strong>of</strong> body fat, <strong>and</strong> fat cell size in the lean rats were<br />

observed (107). In the obese rats food intake <strong>and</strong> body weight<br />

were reduced, but the percent <strong>of</strong> body fat remained unchanged<br />

<strong>and</strong> maintained a fat cell size equivalent to that <strong>of</strong> their obese<br />

controls. This study indicates that the obese rats, despite a<br />

substantial reduction in body weight produced by (-)-<strong>HCA</strong>,<br />

still defend their obese body <strong>com</strong>position. Rao <strong>and</strong> Sakariah<br />

(108) observed that inclusion <strong>of</strong> (-)-<strong>HCA</strong> in the lipogenic diet<br />

resulted in significant reduction in food intake, body weight,<br />

epididymal fat, <strong>and</strong> serum triglyceride in the albino rats <strong>and</strong><br />

also a decrease in the feed efficiency ratio. The decreases in<br />

food intake, body-weight gain, <strong>and</strong> feed efficiency ratio brought<br />

about by (-)-<strong>HCA</strong> were dependent on the content <strong>of</strong> this<br />

<strong>com</strong>pound in the diet. The study encouraged exploration <strong>of</strong> the<br />

efficacy <strong>of</strong> (-)-<strong>HCA</strong> in the control <strong>of</strong> obesity <strong>and</strong> hypertriglyceridemia.<br />

Effect <strong>of</strong> ())-<strong>HCA</strong> on Ketogenesis. Brunengraber et al. (95)<br />

reported that ketogenesis by livers <strong>of</strong> fed rats perfused without<br />

free fatty acid is strongly inhibited by (-)-<strong>HCA</strong>. It seems that<br />

such ketogenesis occurs via an extramitochondrial pathway that<br />

depends on the citrate cleavage reaction for its supply <strong>of</strong><br />

extramitochondrial acetyl-CoA, which operates in carbohydratefed<br />

animals (109). However, the same parameter was not<br />

inhibited by (-)-<strong>HCA</strong> when livers from starved rats were<br />

perfused with oleate. On the other h<strong>and</strong>, the ketogenesis<br />

increased somewhat by (-)-<strong>HCA</strong> when livers from fed rats were<br />

perfused with oleate. The intramitochondrial pathway predominates<br />

either in starved animals or when the concentration <strong>of</strong><br />

fatty acid is high or both. In the case <strong>of</strong> the rumen epithelium<br />

<strong>of</strong> sheep, the absence <strong>of</strong> any significant cytoplasmic <strong>com</strong>ponent<br />

was emphasized by (-)-<strong>HCA</strong>, which should inhibit ketogenesis<br />

if ATP:citrate lyase plays any function in the process (110),<br />

but there was no such inhibition, lending support to the theory<br />

that ketogenesis proceeds exclusively through the mitochondrial<br />

pathway.<br />

Other Biological Effects <strong>of</strong> ())-<strong>HCA</strong>. (-)-<strong>HCA</strong> did not<br />

affect the rate <strong>of</strong> oxygen consumption by rat brain synaptosomes,<br />

<strong>and</strong> the activities <strong>of</strong> fatty acid synthase, carnitine acetyltransferase,<br />

glucose 6-phosphate-dehydrogenase, <strong>and</strong> acetyl-CoA<br />

synthetase but inhibited the activities <strong>of</strong> isocitrate dehydrogenase,<br />

malate dehydrogenase (decarboxylating), <strong>and</strong> aconitate<br />

hydratase at millimolar concentrations (50). (-)-<strong>HCA</strong> blocked<br />

dexamethasone stimulation <strong>of</strong> cytidylyltransferase but not <strong>of</strong><br />

fatty acid synthase in lung tissue (111).<br />

Brunengraber et al. (95) observed that in rat liver, the<br />

inhibition <strong>of</strong> fatty acid synthesis by (-)-<strong>HCA</strong> was associated<br />

with an increase in the tissue content <strong>of</strong> glucose 6-phosphate<br />

<strong>and</strong> fructose 6-phosphate <strong>and</strong> a diminution in glycolytic<br />

intermediates from fructosebisphosphate to phosphoenol pyruvate.<br />

Presumably, the citrate content is elevated in cytoplasm<br />

in the presence <strong>of</strong> (-)-<strong>HCA</strong>. This can be expected to result in<br />

a reduced activity <strong>of</strong> phosph<strong>of</strong>ructokinase because citrate is wellknown<br />

to act as an inhibitor <strong>of</strong> this enzyme (112-114). It has<br />

also been seen that AMP contents drop in the presence <strong>of</strong> (-)-<br />

<strong>HCA</strong>. This can also be expected to reduce the activity <strong>of</strong><br />

phosph<strong>of</strong>ructokinase, because AMP is an activator <strong>of</strong> this<br />

enzyme (115). The inhibition <strong>of</strong> phosph<strong>of</strong>ructokinase by (-)-<br />

<strong>HCA</strong> in rat hepatocytes has also been reported by McCune et<br />

al. (116). It can be concluded that in rat liver, the inhibition <strong>of</strong><br />

phosph<strong>of</strong>ructokinase by (-)-<strong>HCA</strong> leads to the accumulation <strong>of</strong><br />

glucose 6-phosphate <strong>and</strong> fructose 6-phosphate <strong>and</strong> the decrease<br />

<strong>of</strong> glycolytic intermediates beyond fructosebisphosphate as the<br />

reaction catalyzed by phosph<strong>of</strong>ructokinase in glycolysis is<br />

irreversible <strong>and</strong> controls the glycolysis.<br />

Chronic metabolic acidosis increases proximal tubular citrate<br />

uptake, causing hypocitraturia associated with an increase in<br />

cortical ATP:citrate lyase activity <strong>and</strong> protein abundance.<br />

Hypokalemia, which causes only intracellular acidosis, also<br />

caused hypocitraturia <strong>and</strong> increased renal cortical ATP:citrate<br />

lyase activity. Inhibition <strong>of</strong> this enzyme with (-)-<strong>HCA</strong> significantly<br />

abated hypocitraturia <strong>and</strong> increased urinary citrate excretion<br />

in chronic metabolic acidosis <strong>and</strong> in K + depletion (117).<br />

These results suggest an important role <strong>of</strong> ATP:citrate lyase in<br />

proximal tubular citrate metabolism. (-)-<strong>HCA</strong> has been reported<br />

to alter the pyruvate metabolism by tumorigenic cells (118).<br />

Pyruvate consumption by tumor cells declined, but the mean<br />

percentage <strong>of</strong> oxidation increased with (-)-<strong>HCA</strong>.<br />

Possible Mode <strong>of</strong> Action on Reduction in Food Intake.<br />

The chronic oral administration <strong>of</strong> (-)-<strong>HCA</strong> to growing rats<br />

caused a reduction in body-weight gain, food consumption, <strong>and</strong><br />

total body lipids. However, administration <strong>of</strong> equimolar amounts<br />

<strong>of</strong> citrate did not alter weight gain, appetite, or body lipids, <strong>and</strong><br />

there was no increase in liver size or liver lipid content with<br />

either treatment. Paired feeding studies demonstrated that the<br />

reduction in food intake accounted for the decrease in weight<br />

gain <strong>and</strong> body lipid observed with (-)-<strong>HCA</strong> treatment, <strong>and</strong> this<br />

decrease in calorie intake was not responsible for the druginduced<br />

depression <strong>of</strong> hepatic lipogenesis (66, 67). Rao <strong>and</strong><br />

Sakariah (108), in their studies, suggested that the reduction in<br />

appetite in (-)-<strong>HCA</strong>-fed rats seems to be a specific effect <strong>of</strong><br />

(-)-<strong>HCA</strong> ingestion <strong>and</strong> not due to alterations <strong>of</strong> taste as the<br />

diet fed to control group contained citrate at a level equivalent<br />

to that <strong>of</strong> (-)-<strong>HCA</strong> in the experimental diet. In view <strong>of</strong> the<br />

close structural relationship <strong>of</strong> (-)-<strong>HCA</strong> to citrate, it is<br />

reasonable to believe that (-)-<strong>HCA</strong> does not affect the taste <strong>of</strong><br />

the food. Panksepp et al. (119) evaluated the capacity <strong>of</strong> various<br />

salts <strong>of</strong> (-)-<strong>HCA</strong> to produce conditioned rejection <strong>of</strong> a 0.25%<br />

saccharin solution <strong>and</strong> concluded that the food intake was<br />

reduced by (-)-<strong>HCA</strong> only during the first hour following<br />

administration <strong>of</strong> the drug <strong>and</strong> the magnitude <strong>of</strong> appetite<br />

rejection did not correspond to the degree <strong>of</strong> conditioned<br />

rejection, lending support to the conclusion that the food intake<br />

reduction was not merely a consequence <strong>of</strong> aversive effects <strong>of</strong><br />

the drug.<br />

A number <strong>of</strong> <strong>com</strong>plex factors have been implicated in the<br />

appetite regulation <strong>and</strong> feeding behavior (67). Glucose utilization<br />

rates (120), unidentified humoral factor(s) in blood from satiated<br />

rats (121), enterogastrone (122) <strong>and</strong> other gastrointestinal<br />

hormones, plasma <strong>and</strong> brain tryptophan, brain serotonin interrelationships<br />

(123-127), <strong>and</strong> brain catecholamines (128) have<br />

all been suggested to play their role in the regulation <strong>of</strong> feeding<br />

behavior. Because (-)-<strong>HCA</strong> was demonstrated to inhibit fatty<br />

acid <strong>and</strong> cholesterol synthesis, presumably through a reduction


16 J. Agric. Food Chem., Vol. 50, No. 1, 2002 Reviews<br />

in acetyl-CoA pool in lipogenic tissues (65), the same effect<br />

would be expected in any cell possessing ATP:citrate lyase.<br />

Ricny <strong>and</strong> Tucek (129) have observed that the availability <strong>of</strong><br />

acetyl-CoA had a decisive influence on both the rate <strong>of</strong> synthesis<br />

<strong>of</strong> acetylcholine <strong>and</strong> its steady-state concentration with glucose<br />

as the metabolic substrate in rat brain slices. Acetylcholine<br />

released by the phrenic nerve was measured in the isolated<br />

perfused rat hemidiaphragm; when (-)-<strong>HCA</strong> was added to the<br />

perfusate, the release <strong>of</strong> acetylcholine was stabilized at a level<br />

40% below the control value (130). (-)-<strong>HCA</strong> was reported to<br />

inhibit the synthesis <strong>of</strong> acetylcholine in rat brain slices (90, 131,<br />

132). These results suggested that citrate transport <strong>and</strong> subsequent<br />

cleavage give rise to acetyl-CoA as the principal precursor<br />

<strong>of</strong> acetylcholine synthesis. Sterling et al. (133) reported that in<br />

rat brain cortex, (-)-<strong>HCA</strong> reduced the incorporation <strong>of</strong> [ 3 H]-<br />

glucose <strong>and</strong> [ 14 C]-β-hydroxybutyrate to the acetyl moiety <strong>of</strong><br />

acetylcholine. They suggested that in adult rats, β-hydroxybutyrate<br />

could contribute to the acetyl moiety <strong>of</strong> acetylcholine,<br />

possibly via the citrate cleavage pathway. Ricny <strong>and</strong> Tucek (134)<br />

observed the effects <strong>of</strong> (-)-<strong>HCA</strong> on the concentration <strong>of</strong> acetyl-<br />

CoA <strong>and</strong> acetylcholine in the tissue <strong>and</strong> on the release <strong>of</strong><br />

acetylcholine into the media were investigated in experiments<br />

on slices <strong>of</strong> rat caudate nuclei incubated in different media. With<br />

glucose as the main metabolic substrate, (-)-<strong>HCA</strong> diminished<br />

the concentration <strong>of</strong> acetyl-CoA in all <strong>of</strong> the media used. ATP:<br />

citrate lyase appears to provide about one-third <strong>of</strong> acetyl-CoA<br />

used for the synthesis <strong>of</strong> acetylcholine. Gibson <strong>and</strong> Peterson<br />

(135) have shown that the (-)-<strong>HCA</strong>-reduced incorporation <strong>of</strong><br />

[U- 14 C]glucose into acetylcholine was greater in septal than in<br />

hippocampal slices, indicating that acetylcholine metabolism<br />

varies regionally. Recently, Szutowicz et al. (136) reported that<br />

in Ca 2+ -activated rat synaptosomes, both synaptosomic acetyl-<br />

CoA <strong>and</strong> acetylcholine syntheses were suppressed by 27 <strong>and</strong><br />

29%, respectively, by 1 mM (-)-<strong>HCA</strong>. The impairment <strong>of</strong><br />

acetylcholine synthesis with (-)-<strong>HCA</strong> was also reported in the<br />

brain stem <strong>of</strong> neonatal rat (137). All <strong>of</strong> these in vitro studies<br />

revealed that (-)-<strong>HCA</strong> can depress acetylcholine synthesis in<br />

nerve tissues, although so far there is no evidence <strong>of</strong> reduced<br />

synthesis <strong>of</strong> acetylcholine in intact animals with the administration<br />

<strong>of</strong> (-)-<strong>HCA</strong>. Sullivan et al. (67) suggested that if (-)-<br />

<strong>HCA</strong> could penetrate the blood-brain barrier, then the depression<br />

<strong>of</strong> acetylcholine levels or rate <strong>of</strong> turnover in the brain<br />

resulting from a decreased precursor pool could affect cholinergic<br />

receptor systems that might be involved in feeding<br />

behavior.<br />

Fatty acid synthesis regulates fatty acid oxidation by a wellcharacterized<br />

mechanism (138, 139). In this paradigm, malonyl-<br />

CoA levels rise during fatty acid synthesis <strong>and</strong> result in<br />

inhibition <strong>of</strong> carnitine palmitoyl transferase 1 (CPT 1) mediated<br />

uptake <strong>of</strong> fatty acids into the mitochondria. This results in<br />

elevation <strong>of</strong> cytoplasmic long-chain fatty acyl (LCFA)-CoAs<br />

<strong>and</strong> diacylglycerol molecules that may play a role in signaling,<br />

which leads to the proposal that malonyl-CoA levels act as a<br />

signal <strong>of</strong> the availability <strong>of</strong> physiological fuels (140). One such<br />

proposal for maolnyl-CoA is the mediation <strong>of</strong> nutrient-stimulated<br />

secretion in the beta cells. Glucose-sensing neurons, which<br />

regulate feeding in the hypothalamus, share many features with<br />

the beta cells, including expression <strong>of</strong> glucokinase <strong>and</strong> the<br />

adenosine-sensitive potassium channel (141). The studies <strong>of</strong><br />

Chen et al. (142) showed that (-)-<strong>HCA</strong> pr<strong>of</strong>oundly inhibited<br />

the glucose-stimulated insulin secretion (GSIS) from beta cells<br />

<strong>and</strong> also provided evidence for the pivotal role <strong>of</strong> malonyl-CoA<br />

suppression <strong>of</strong> CPT 1 with attending elevation <strong>of</strong> the cytosolic<br />

LCFA-CoA concentration in GSIS from the normal pancreatic<br />

beta cell. Moreover, Saha et al. (103) reported that inhibition<br />

<strong>of</strong> ATP:citrate lyase by (-)-<strong>HCA</strong> markedly diminished the<br />

malonyl-CoA level, indicating that citrate was the major<br />

substrate for the malonyl-CoA precursor, that is, cytosolic acetyl-<br />

CoA, <strong>and</strong> also there is sufficient evidence that (-)-<strong>HCA</strong> inhibits<br />

ATP:citrate lyase (47-51), limiting the pool <strong>of</strong> cytosolic acetyl-<br />

CoA, the precursor <strong>of</strong> malonyl-CoA. This type <strong>of</strong> regulation <strong>of</strong><br />

malonyl-CoA level may affect the signaling <strong>of</strong> fuel status in<br />

hypothalamic neurons regulating feeding behavior, lending<br />

support that (-)-<strong>HCA</strong> may represent a biochemical target for<br />

the control <strong>of</strong> appetite/feeding behavior <strong>and</strong> body weight. Thus,<br />

(-)-<strong>HCA</strong> seems to act at the metabolic level <strong>and</strong> not directly<br />

at the central nervous system as classical appetite depressants<br />

do.<br />

())-<strong>HCA</strong> May Promote Glycogenesis, Gluconeogenesis,<br />

<strong>and</strong> Lipid Oxidation. The inhibition <strong>of</strong> ATP:citrate lyase by<br />

(-)-<strong>HCA</strong> causes less dietary carbohydrate to be utilized for the<br />

synthesis <strong>of</strong> fatty acids, resulting in more glycogen storage in<br />

the liver <strong>and</strong> muscles. Sullivan et al. (143) reported an increase<br />

in the rate <strong>of</strong> in vivo hepatic glycogen synthesis with the<br />

administration <strong>of</strong> (-)-<strong>HCA</strong>. Sullivan et al. (144) <strong>and</strong> Sullivan<br />

<strong>and</strong> Green (145) proposed that (-)-<strong>HCA</strong> acted primarily through<br />

its effects on the appetite, possibly involving increases in<br />

glycogen levels. Even though the administration <strong>of</strong> (-)-<strong>HCA</strong><br />

increased the hepatic glycogen contents in rats, Hellerstein <strong>and</strong><br />

Xie (146) demonstrated that neither hepatic glycogen nor the<br />

hexosephosphates are involved in the food-intake suppressive<br />

effects <strong>of</strong> (-)-<strong>HCA</strong>. It seems the increased hepatic fatty acid<br />

oxidation leading to increased levels <strong>of</strong> acetyl-CoA <strong>and</strong> ATP<br />

plays some role in this effect.<br />

In a well-fed state, the hepatic carnitine levels are far too<br />

low to activate CPT 1. The rate-limiting enzyme for hepatic<br />

lipid oxidation, CPT 1, is activated by exogenous carnitine <strong>and</strong><br />

inhibited by malonyl-CoA. The lipogenesis inhibitor (-)-<strong>HCA</strong><br />

can decrease the production <strong>of</strong> malonyl-CoA in hepatocytes by<br />

potent inhibition <strong>of</strong> ATP:citrate lyase <strong>and</strong> may activate CPT 1,<br />

which can facilitate lipid oxidation. Earlier studies demonstrated<br />

that (-)-<strong>HCA</strong> can reduce body-weight <strong>and</strong> fat accumulation in<br />

growing rats, owing to the reduction in appetite. From the above<br />

views McCarty (147) hypothesized that joint administration <strong>of</strong><br />

(-)-<strong>HCA</strong> <strong>and</strong> carnitine should therefore promote hepatic lipid<br />

oxidation, gluconeogenesis, <strong>and</strong> satiety. In a recent study, joint<br />

administration <strong>of</strong> pyruvate, (-)-<strong>HCA</strong>, <strong>and</strong> carnitine to obese<br />

subjects was associated with a remarkable rate <strong>of</strong> body-fat loss<br />

<strong>and</strong> thermogenesis, strongly suggestive <strong>of</strong> uncoupled fatty acid<br />

oxidation (148). The reduction <strong>of</strong> free fatty acids by stimulating<br />

hepatic oxidation with (-)-<strong>HCA</strong> <strong>and</strong> carnitine may ameliorate<br />

risk factors associated with abdominal obesity (149).<br />

Sustained aerobic exercise requires a severalfold increase in<br />

hepatic glucose output. An increasing proportion <strong>of</strong> this elevated<br />

glucose output must be provided by gluconeogenesis. Thus,<br />

preadministration <strong>of</strong> (-)-<strong>HCA</strong> may aid endurance during<br />

postabsorptive aerobic exercise by promoting gluconeogenesis.<br />

Carnitine <strong>and</strong> bioactive chromium may potentiate this benefit.<br />

The utility <strong>of</strong> this technique may be greatest in exercise regimens<br />

designed to promote weight loss (150). The ability <strong>of</strong> exercise<br />

to selectively promote fat oxidation should be optimized if<br />

exercise is done postabsorptively (preferably during morning<br />

fasting metabolism), if (-)-<strong>HCA</strong> <strong>and</strong> carnitine are administered<br />

prior to exercise, if the exercise regimen is <strong>of</strong> moderate intensity<br />

<strong>and</strong> prolonged duration, <strong>and</strong> if no calories are ingested for<br />

several hours following exercise (151).<br />

Kriketos et al. (152) did not detect any effect <strong>of</strong> (-)-<strong>HCA</strong><br />

administration on lipid oxidation, respiratory quotient (RQ), <strong>and</strong>


Reviews J. Agric. Food Chem., Vol. 50, No. 1, 2002 17<br />

energy expenditure in humans during either exercise or moderately<br />

intense exercise. However, Ishihara et al. (153) demonstrated<br />

that chronic administration <strong>of</strong> (-)-<strong>HCA</strong> to mice<br />

lowered the RQ <strong>and</strong> increased lipid oxidation during both resting<br />

<strong>and</strong> exercising conditions <strong>and</strong> increased the endurance in<br />

exercise. These results indicated that the enhancement <strong>of</strong><br />

endurance exercise by chronically administered (-)-<strong>HCA</strong> in<br />

mice might have occurred by the attenuation <strong>of</strong> glycogen<br />

consumption caused by the promotion <strong>of</strong> lipid oxidation during<br />

exercise.<br />

(-)-<strong>HCA</strong> AS A WEIGHT-CONTROLLING AGENT<br />

Weight gain occurs when the limited capacity for storing<br />

glycogen in the liver <strong>and</strong> muscles is attained, <strong>and</strong> beyond this<br />

point excess glucose is converted into fat <strong>and</strong> stored in fat cells<br />

throughout the body. (-)-<strong>HCA</strong> exerts its antiobesity effect by<br />

inhibiting ATP:citrate lyase, consequently inhibiting the cleavage<br />

<strong>of</strong> citrate to oxaloacetate <strong>and</strong> acetyl-CoA, a key molecule, which<br />

plays a critical role in energy storage as fat. Now, instead <strong>of</strong><br />

wasting energy to synthesize fat, the energy is diverted to the<br />

production <strong>of</strong> glycogen in the liver <strong>and</strong> muscles. This slows<br />

the production <strong>of</strong> fatty acids, cholesterol, <strong>and</strong> triglycerides with<br />

the net effect <strong>of</strong> reduced fat production <strong>and</strong> storage (154).<br />

Preliminary research based on laboratory <strong>and</strong> animal experiments<br />

suggests that (-)-<strong>HCA</strong> may be a useful weight loss aid<br />

(101, 102). (-)-<strong>HCA</strong> has been demonstrated in the laboratory<br />

to reduce the conversion <strong>of</strong> carbohydrates into stored fat by<br />

inhibiting certain enzyme processes (49, 65). Animal research<br />

also indicated that (-)-<strong>HCA</strong> suppresses appetite <strong>and</strong> food intake<br />

to induce weight loss (66, 67, 104, 107, 108).<br />

Heymsfield et al. (155) stated in their publication that<br />

although (-)-<strong>HCA</strong> appears to be a promising experimental<br />

weight control agent, studies in humans are limited (156-160)<br />

<strong>and</strong> results have been contradictory. Supporting evidence <strong>of</strong><br />

human (-)-<strong>HCA</strong> efficacy for weight control is based largely<br />

on studies with small sample sizes (157, 158), studies that failed<br />

to include a placebo-treated group (156), <strong>and</strong> use <strong>of</strong> inaccurate<br />

measures <strong>of</strong> body lipid change (158). Even though the effectiveness<br />

<strong>of</strong> (-)-<strong>HCA</strong> is unclear, at least 14 separate (-)-<strong>HCA</strong>containing<br />

products are sold over-the-counter to consumers<br />

(161). To over<strong>com</strong>e limitations <strong>of</strong> the above studies <strong>and</strong> examine<br />

the effectiveness <strong>of</strong> (-)-<strong>HCA</strong> for weight loss <strong>and</strong> fat mass<br />

reduction, Heymsfield <strong>and</strong> his colleagues at Obesity Research<br />

Center, University College <strong>of</strong> Physicians <strong>and</strong> Surgeons, New<br />

York, designed their investigation with rigorous controlled trials.<br />

Subjects were r<strong>and</strong>omized to receive either active herbal<br />

<strong>com</strong>pound [1500 mg <strong>of</strong> (-)-<strong>HCA</strong> per day] or placebo. Both<br />

groups were prescribed a high-fiber, low-energy diet, <strong>and</strong> the<br />

treatment period was 12 weeks. Body weight was evaluated<br />

every week, <strong>and</strong> fat mass was measured at weeks 0 <strong>and</strong> 12. On<br />

the basis <strong>of</strong> the results obtained from these experiments,<br />

Heymsfield et al. (155) concluded that a prospective doubleblind<br />

study failed to detect either weight loss or fat-mobilizing<br />

effects <strong>of</strong> (-)-<strong>HCA</strong> beyond those <strong>of</strong> a placebo.<br />

Rebuttal letters from Vladimir Badmaev et al. <strong>of</strong> Sabinsa<br />

Corp. (a manufacturer <strong>of</strong> st<strong>and</strong>ardized G. cambogia extract);<br />

James L. Schaller, Birchrrunville, PA; <strong>and</strong> Fabio Firenzuoli <strong>and</strong><br />

Luigi Gori, St. Joseph Hospital, Empoli, Italy, published in the<br />

July 21, 1999, issue <strong>of</strong> the Journal <strong>of</strong> the American Medical<br />

Association were critical <strong>of</strong> Heymsfield’s studies. The letters<br />

are summarized below:<br />

• The results are contradictory to the positive results reported<br />

in several (-)-<strong>HCA</strong> clinical studies (156-160, 162), <strong>and</strong> it<br />

cannot be concluded from this single study that (-)-<strong>HCA</strong> is<br />

ineffective.<br />

• Previous studies <strong>and</strong> their own preclinical research (163)<br />

suggest that for (-)-<strong>HCA</strong> to effectively inhibit fat formation<br />

<strong>and</strong> body-weight gain, it needs to be administered with a simple<br />

carbohydrate-rich (lipogenic) diet.<br />

• The co-administration <strong>of</strong> (-)-<strong>HCA</strong> with a high-fiber diet<br />

may have inhibited the gastrointestinal absorption <strong>of</strong> (-)-<strong>HCA</strong>.<br />

This issue be<strong>com</strong>es critical with (-)-<strong>HCA</strong>; its reported efficacy<br />

in inhibiting intracellular enzyme ATP:citrate lyase depends<br />

entirely on the presence <strong>of</strong> (-)-<strong>HCA</strong> inside the target cell. The<br />

significance <strong>of</strong> (-)-<strong>HCA</strong> availability in the cytosol <strong>of</strong> the target<br />

cell for inhibiting lipid synthesis or ATP citrate lyase was<br />

confirmed (164).<br />

• The low-energy diet used in the study by Heymsfield et al.<br />

negated the utility <strong>of</strong> (-)-<strong>HCA</strong>. The study <strong>of</strong> Heymsfield et al.<br />

failed to evaluate (-)-<strong>HCA</strong>’s ability to reduce food intake by<br />

virtue <strong>of</strong> its energy-restricted protocol.<br />

• The dose <strong>of</strong> (-)-<strong>HCA</strong> used in the study was low <strong>com</strong>pared<br />

with that used in earlier animal studies (66, 67).<br />

• Excess calcium used to stabilize the (-)-<strong>HCA</strong> molecule,<br />

<strong>com</strong>mon among products, reduces solubility <strong>and</strong> hinders bioavailability.<br />

A soluble Garcinia powder <strong>and</strong> liquid extract<br />

containing a lactone from (-)-<strong>HCA</strong> was <strong>com</strong>pared with a<br />

calcium-type Garcinia powder administered in food to rats, <strong>and</strong><br />

the liquid extract was proven to be more effective (165). Because<br />

blood (-)-<strong>HCA</strong> levels were not measured in the study, it was<br />

not known whether bioavailability was ensured in the studies<br />

<strong>of</strong> Heymsfield et al. (155).<br />

SOME POSSIBLE CONCERNS ABOUT (-)-<strong>HCA</strong><br />

Animal studies indicate that (-)-<strong>HCA</strong> is no more toxic than<br />

citric acid itself, which is present in many foods in addition to<br />

being a normal intracellular <strong>com</strong>pound. Also, (-)-<strong>HCA</strong> is a<br />

<strong>com</strong>ponent <strong>of</strong> a natural product, which has been used in Indian<br />

cuisine as well as for medicinal purposes (1). Clouatre <strong>and</strong><br />

Rosenbaum (154) pointed out that (-)-<strong>HCA</strong> has extremely low<br />

levels <strong>of</strong> toxicity. For example, recent oral toxicity studies<br />

performed at Wil Research Laboratories in Ashl<strong>and</strong>, OH, found<br />

that 5000 mg/kg <strong>of</strong> body weight <strong>of</strong> (-)-<strong>HCA</strong> resulted in no<br />

visible symptoms <strong>of</strong> toxicity or deaths in laboratory animals.<br />

This is roughly equivalent to 350 g or 233 times the dosage <strong>of</strong><br />

1.5 g/day <strong>of</strong> (-)-<strong>HCA</strong> that might be consumed by an averagesized<br />

person. Because G. cambogia has a long history <strong>of</strong> usage<br />

as a flavoring agent, preservative, <strong>and</strong> herbal tonic <strong>and</strong> there<br />

are no reports <strong>of</strong> toxicity regarding traditional use <strong>of</strong> the<br />

Garcinia extract, it is highly unlikely that there may be any<br />

possible negative effect that may occur due to excess intake;<br />

the possibility <strong>of</strong> bowel intolerance can be easily reversed by<br />

simply reducing the dosage. However, this problem has not been<br />

reported in animal studies at the levels that were necessary to<br />

reduce appetite. However, despite its inherent safety, (-)-<strong>HCA</strong>,<br />

like any other diet product, is not re<strong>com</strong>mended for certain<br />

groups <strong>of</strong> people. (-)-<strong>HCA</strong> has impacts on the body’s production<br />

<strong>of</strong> fatty acids <strong>and</strong> cholesterol; therefore, it may directly<br />

influence the production <strong>of</strong> sterols, thus restricting the production<br />

<strong>of</strong> steroid hormones. As pregnancy is a time <strong>of</strong> extreme<br />

sensitivity to steroid hormones, products containing (-)-<strong>HCA</strong><br />

should not be re<strong>com</strong>mended during pregnancy. Likewise,<br />

women who are breast-feeding should also avoid (-)-<strong>HCA</strong>.<br />

Although experience with fruit sources <strong>of</strong> (-)-<strong>HCA</strong> shows that<br />

they are not dangerous to young children, they are advised not<br />

to consume (-)-<strong>HCA</strong> in large amounts for extended periods.<br />

One concern is about the time <strong>of</strong> administration <strong>and</strong> dosage<br />

considerations <strong>of</strong> (-)-<strong>HCA</strong> for its efficacy. Sullivan et al. (65)<br />

demonstrated that the effects <strong>of</strong> (-)-<strong>HCA</strong> in animals depend


18 J. Agric. Food Chem., Vol. 50, No. 1, 2002 Reviews<br />

on the time <strong>of</strong> administration in relation to a meal, with (-)-<br />

<strong>HCA</strong> maximally effective when administered 30-60 min prior<br />

to feeding. Most <strong>of</strong> the clinical studies including that <strong>of</strong><br />

Heymsfield et al. (155) administered (-)-<strong>HCA</strong> ∼30 min prior<br />

to meals, which is the lower end <strong>of</strong> the maximally effective<br />

range. However, the administration <strong>of</strong> (-)-<strong>HCA</strong> postabsorptively<br />

in the studies <strong>of</strong> Kriketos et al. (152) showed a trend<br />

toward a lower RQ during (-)-<strong>HCA</strong> phase, although it was<br />

not statistically significant. Even though this study did not<br />

support the hypothesis that (-)-<strong>HCA</strong> alters the short-term rate<br />

<strong>of</strong> fat oxidation, the data certainly do not rule out its possibility.<br />

Another related concern is that (-)-<strong>HCA</strong> provided in divided<br />

doses was found to be more effective than the same amount<br />

given as single dose (67). In weight-loss protocols human doses<br />

ranging between 750 <strong>and</strong> 1500 mg/day <strong>of</strong> (-)-<strong>HCA</strong> (156-160)<br />

are the extreme lower end <strong>of</strong> the in vivo dose response range<br />

established by Sullivan et al. (66).<br />

As discussed earlier, the administration <strong>of</strong> (-)-<strong>HCA</strong> increases<br />

the rate <strong>of</strong> gluconeogenesis <strong>and</strong> ketone bodies formation (166),<br />

which can be deleterious in subjects with type-2 diabetes. In<br />

this situation, we can consider the hypothesis <strong>of</strong> McCarty (167)<br />

that excessive exposure <strong>of</strong> tissues to fatty acids is likely to be<br />

the chief cause <strong>of</strong> the various dysfunctions that lead to sustained<br />

hyperglycemia in type-2 diabetes. Disinhibition <strong>of</strong> hepatic fatty<br />

acid oxidation <strong>and</strong> inhibition <strong>of</strong> fatty acid synthesis with (-)-<br />

<strong>HCA</strong> <strong>and</strong> carnitine, as suggested earlier (147), have considerable<br />

potential as a new weight-loss strategy, but diabetics run the<br />

risk <strong>of</strong> further enhancing excessive hepatic gluconeogenesis.<br />

McCarty (147) hypothesized that the clinical utility <strong>of</strong> metformin<br />

in diabetes is probably traceable to inhibition <strong>of</strong> gluconeogenesis,<br />

<strong>and</strong> its use as an adjunct to (-)-<strong>HCA</strong>/carnitine treatment<br />

<strong>of</strong> obesity in diabetics deserves evaluation, particularly as<br />

metformin therapy itself tends to reduce body weight. Furthermore,<br />

metformin therapy will not impede the activation <strong>of</strong> fatty<br />

acid oxidation by (-)-<strong>HCA</strong>/carnitine <strong>and</strong> is likely to potentiate<br />

the appetite-suppressant <strong>and</strong> thermogenic benefits <strong>of</strong> this<br />

strategy. Indeed, because metformin has been reported to lower<br />

body weight <strong>and</strong> improve cardiovascular risk factors in obese<br />

nondiabetics, a broader application <strong>of</strong> an (-)-<strong>HCA</strong>/carnitine/<br />

metformin therapy for obesity can be contemplated.<br />

(-)-<strong>HCA</strong> may have deleterious effects on insulin sensitivity.<br />

Many researchers have suggested that malonyl-CoA has an<br />

important role in transmitting the insulin signal in the cells (140,<br />

168, 169). It is well-known that (-)-<strong>HCA</strong> prevents the production<br />

<strong>of</strong> acetyl-CoA <strong>and</strong> subsequently malonyl-CoA (103);<br />

therefore, preventing its production may impair insulin responsiveness<br />

in some (or all) tissues. Thus, (-)-<strong>HCA</strong> is likely to<br />

have some effect on insulin sensitivity that can lead to type-2<br />

diabetes, that is, NIDDM.<br />

A role <strong>of</strong> malonyl-CoA in glucose-stimulated insulin secretion<br />

(GSIS) has been defined <strong>and</strong> a rise in malonyl-CoA, which<br />

preceded the insulin secretion in clonal pancreatic beta cells,<br />

was evident after exposure to glucose (170). Sener <strong>and</strong> Malaisse<br />

(171) reported the failure <strong>of</strong> (-)-<strong>HCA</strong> in the range <strong>of</strong> 1.0-2.0<br />

mM to affect the GSIS in glucose-stimulated intact pancreatic<br />

islets <strong>of</strong> rat. However, Chen et al. (142) explored the emerging<br />

concept <strong>of</strong> Prentki et al. (172) that malonyl-CoA generation with<br />

con<strong>com</strong>itant suppression <strong>of</strong> mitochondrial CPT 1 represents an<br />

important <strong>com</strong>ponent <strong>of</strong> GSIS by pancreatic beta cells <strong>and</strong><br />

observed that (-)-<strong>HCA</strong> pr<strong>of</strong>oundly inhibited GSIS <strong>and</strong> also<br />

provided more direct evidence for a pivotal role <strong>of</strong> malonyl-<br />

CoA suppression <strong>of</strong> CPT 1 with attendant elevation <strong>of</strong> the<br />

cytosolic long-chain acyl-CoA concentration in GSIS from the<br />

normal pancreatic beta cell. There may be another advantage<br />

<strong>of</strong> (-)-<strong>HCA</strong> in that it inhibits GSIS, which helps to keep this<br />

fat-storing hormone in check.<br />

High blood sugar levels suppress the burning <strong>of</strong> fatty acids<br />

for the availability <strong>of</strong> energy. Within the past few years,<br />

researchers have shown that high blood sugar levels generate<br />

malonyl-CoA, which is an inhibitor <strong>of</strong> carnitine palmitoyl<br />

transferase (CPT 1) that controls the transfer <strong>of</strong> long-chain fatty<br />

acyl (LCFA)-CoA into the mitochondria where these are<br />

oxidized. An increase in malonyl-CoA also results in the<br />

elevation <strong>of</strong> acyl-CoA, which triggers the release <strong>of</strong> insulin (140,<br />

168, 169, 173-176). Saha et al. (103) observed that inhibition<br />

<strong>of</strong> ATP:citrate lyase with (-)-<strong>HCA</strong> markedly diminished the<br />

increase in malonyl-CoA in skeletal muscle, indicating that<br />

citrate was the major substrate for malonyl-CoA precursor <strong>and</strong><br />

cytosolic acetyl-CoA. As CPT 1 is considered to be a ratelimiting<br />

factor in the burning <strong>of</strong> fatty acids (177), the trick <strong>of</strong><br />

long-term permanent fat loss is to increase the activity <strong>of</strong> CPT<br />

1. (-)-<strong>HCA</strong> can be used to increase the activity <strong>of</strong> CPT 1 as<br />

does L-carnitine <strong>and</strong> glucagon. Because research has shown that<br />

the conversion <strong>of</strong> carbohydrates into fat is prevented by (-)-<br />

<strong>HCA</strong>, a more important function <strong>of</strong> this incredible nutrient is<br />

its ability to increase the CPT 1 activity by decreasing the pool<br />

<strong>of</strong> acetyl-CoA, thus reducing the level <strong>of</strong> malonyl-CoA <strong>and</strong><br />

raising the activity <strong>of</strong> CPT 1.<br />

LITERATURE CITED<br />

(1) The Wealth <strong>of</strong> India (Raw Materials); CSIR: New Delhi, India,<br />

1956; Vol. IV, pp 99-108.<br />

(2) Bennet, G. J.; Lee, H. H. Xanthones from Guttiferae. Phytochemistry<br />

1989, 28, 967-998.<br />

(3) Rama Rao, A. V.; Venkataswamy, G.; Yemul, S. S. Xanthochymol<br />

& isoxanthochymol; two polyisoprenylated benzophenoes<br />

from Garcinia xanthochymus. Ind. J. Chem. 1980, 19, 627-<br />

633.<br />

(4) Minami, H.; Kinoshita, M.; Fukuyama, Y.; Kodama, M.;<br />

Yoshizawa, T.; Sugiura, M.; Nakagawa, K.; Tago, H. Antioxidant<br />

xanthones from Garcinia subliptica. Phytochemistry 1994, 36,<br />

501-506.<br />

(5) Lowenstein; J. M. Method <strong>of</strong> treating obesity. U.S. Patent<br />

3764692, 1973.<br />

(6) Guthrie, R. W.; Kierstead, R. W. Hydroxycitric acid derivatives.<br />

U.S. Patent 3993667, 1976.<br />

(7) Guthrie, R. W.; Kierstead, R. W. Hydroxycitric acid derivatives.<br />

U.S. Patent 3993668, 1976.<br />

(8) Guthrie, R. W.; Kierstead, R. W. Hydroxycitric acid derivatives.<br />

U.S. Patent 3994927, 1976.<br />

(9) Guthrie, R. W.; Kierstead, R. W. Hydroxycitric acid derivatives.<br />

U.S. Patent 4005086, 1977.<br />

(10) Guthrie, R. W.; Kierstead, R. W. Hydroxycitric acid derivatives.<br />

U.S. Patent 4006166, 1977.<br />

(11) Guthrie, R. W.; Kierstead, R. W.; Hydroxycitric acid derivatives.<br />

U.S. Patent 4007208, 1977.<br />

(12) Guthrie, R. W.; Kierstead, R. W. Hydroxycitric acid derivatives.<br />

U.S. Patent 4028397, 1977.<br />

(13) Nishida, H. Nutritional regulating food in baked, confectionery.<br />

Jpn. Patent 9294563A2, 1997.<br />

(14) Policappelli, N. E.; Garzone, R.; Russo, C. Dietary supplement.<br />

U.S. Patent 5612039, 1997.<br />

(15) Hastings, C. W.; Barnes, D. J. Weight loss <strong>com</strong>position for<br />

burning <strong>and</strong> reducing synthesis <strong>of</strong> fats. U.S. Patent 5626849,<br />

1997.<br />

(16) Wakat, D. Dietary supplements for the cardiovascular system.<br />

U.S. Patent 6054128, 2000.<br />

(17) Tomi, H.; Tamura, K. Beverage. Jpn. Patent 10004939A2, 1998.<br />

(18) Majeed, M.; Badmaev, V.; Rajendran, R. Potassium hydroxycitrate<br />

for the suppression <strong>of</strong> appetite <strong>and</strong> induction <strong>of</strong> weight<br />

loss. U.S. Patent 5783603, 1998.


Reviews J. Agric. Food Chem., Vol. 50, No. 1, 2002 19<br />

(19) Braswell, A.; Glenn, A.; Aftab, J.; Marina Del Ray. Method for<br />

controlling weight with Hypericum perforatum <strong>and</strong> Garcinia<br />

cambogia. U.S. Patent 5911992, 1999.<br />

(20) Okubo, K. T. Obesity inhibitor. Jpn. Patent 11001431A2, 1999.<br />

(21) Fushiki, T.; Ishihara, K. A.; Takahiko, T. H.; Otsu-shi, S. Athletic<br />

endurance increasing agent in food. WO Patent 9835664A1,<br />

1998.<br />

(22) Yamaguchi; Fumio, Saito; Makoto, Ishikawa; Hiroharu, Kataoka;<br />

Shigehiro, Ariga; Toshiaki, Healthy foods <strong>and</strong> cosmetics. U.S.<br />

Patent 5972357, 1999.<br />

(23) Lewis, Y. S.; Neelakantan, S. (-)-Hydroxycitric acidsThe<br />

principal acid in the fruits <strong>of</strong> Garcinia cambogia. Phytochemistry<br />

1965, 4, 619-625.<br />

(24) Lewis, Y. S. Isolation <strong>and</strong> properties <strong>of</strong> hydroxycitric acid. In<br />

Methods in Enzymology; Colowick, S. P., Kaplan, N. O., Eds.;<br />

Academic Press: New York, 1969; Vol. 13, pp 613-619.<br />

(25) Lowenstein, J. M.; Bruneugraber, H. Hydroxycitrate. In Methods<br />

in Enzymology; Lowenstein, J. M., Ed.; Academic Press: New<br />

York, 1981; Vol. 72, pp 486-497.<br />

(26) Watt, G. Dictionary <strong>of</strong> the Economic Products <strong>of</strong> India;<br />

Periodical Export: Delhi, India, 1972; Vol. III, p 464.<br />

(27) Sreenivasan, A.; Venkataraman, R. Chromatographic detection<br />

<strong>of</strong> the organic constituents <strong>of</strong> Gorikapuli (Garcinia cambogia<br />

Desr.) used in pickling fish. Curr. Sci. 1959, 28, 151-152.<br />

(28) Lewis, Y. S.; Neelakantan, S.; Anjana Murthy, C. Acids in<br />

Garcinia cambogia. Curr. Sci. 1964, 33, 82-83.<br />

(29) Kuriyan, K. I.; P<strong>and</strong>ya, K. C. A note on the main constituents<br />

<strong>of</strong> the dried rind <strong>of</strong> the fruit <strong>of</strong> Garcinia cambogia. J. Ind. Chem.<br />

Soc. 1931, 81, 469.<br />

(30) Palmer, J. K. Determination <strong>of</strong> organic acids by ion-exchange<br />

chromatography. Conn. Agric. Exp. Stn. Bull. 1955, No. 1, 589.<br />

(31) M<strong>of</strong>fett, S. A.; Bh<strong>and</strong>ari, A. K.; Ravindranath, B.; Balasubramanvam,<br />

K. Hydroxycitric acid concentrate <strong>and</strong> food products<br />

prepared therefrom. U.S. Patent 5656316, 1996.<br />

(32) M<strong>of</strong>fett, S. A.; Bh<strong>and</strong>ari, A. K.; Ravindranath, B.; Balasubramanvam,<br />

K. Hydroxycitric acid concentrate <strong>and</strong> food products<br />

prepared therefrom. U.S. Patent 5656314, 1997.<br />

(33) Martius, C.; Maue, R. Preparation, physiological behavior, <strong>and</strong><br />

importance <strong>of</strong> hydroxycitric acid <strong>and</strong> its isomers. Z. Physiol.<br />

Chem. 1941, 269, 33.<br />

(34) Boll, P. M.; Else, S.; Erik, B. Naturally occurring lactones <strong>and</strong><br />

lactames III. The absolute configuration <strong>of</strong> the hydroxycitric acid<br />

lactones, Hibiscus acid <strong>and</strong> Garcinia acid. Acta Chem. Sc<strong>and</strong>.<br />

1969, 23, 286-293.<br />

(35) Glusker, J. P.; Minkin, J. A.; Casciato, C. A.; Soule, F. B.<br />

Absolute configuration <strong>of</strong> the naturally occurring hydroxycitric<br />

acids. Arch. Biochem. Biophys. 1969, 132, 573-577.<br />

(36) Glusker, J. P.; Minkin, J. A.; Casciato, C. A. The structure <strong>and</strong><br />

absolute configuration <strong>of</strong> the calcium salt <strong>of</strong> garcinia acid, the<br />

lactone <strong>of</strong> (-)-hydroxycitric acid. Acta Crystallogr. 1971, B27,<br />

1284-1293.<br />

(37) Stallings, W.; Blount, T. F.; Srere, P. A.; Glusker, J. P. Structural<br />

studies <strong>of</strong> hydroxycitrate <strong>and</strong> their relevance to certain enzymatic<br />

mechanisms. Arch. Biochem. Biophys. 1979, 193, 431-448.<br />

(38) Jayaprakasha, G. K.; Sakariah, K. K. Determination <strong>of</strong> (-)-<br />

hydroxycitric acid in <strong>com</strong>mercial samples <strong>of</strong> Garcinia cambogia<br />

extracts by liquid chromatography using ultraviolet detection.<br />

J. Liq. Chromatogr. Relat. Technol. 2000, 23, 915-923.<br />

(39) Antony, J. I. X.; Josan, P. D.; Shankarnarayana, M. L. Quantitative<br />

analysis <strong>of</strong> (-)-hydroxycitric acid <strong>and</strong> (-)-hydroxycitric acid<br />

lactone in Garcinia fruits <strong>and</strong> Garcinia products. J. Food Sci.<br />

Technol. 1998, 35, 399-402.<br />

(40) AOAC. Official Methods <strong>of</strong> Analysis, 11th ed.; Association <strong>of</strong><br />

Official Analytical Chemists: Washington, DC, 1970.<br />

(41) Jayaprakasha, G. K.; Sakariah, K. K. Determination <strong>of</strong> organic<br />

acids in Garcinia cambogia (Desr.) by HPLC. J. Chromatogr.<br />

A 1998, 806, 337-339.<br />

(42) Jayaprakasha, G. K.; Sakariah, K. K. Determination <strong>of</strong> organic<br />

acids in leaves <strong>and</strong> rinds <strong>of</strong> Garcinia indica (Desr.) by HPLC.<br />

J. Pharm. Biomed. Anal. 2001, in press.<br />

(43) Krishnamurthy, N.; Lewis, Y. S.; Ravindranath, B. Chemical<br />

constituents <strong>of</strong> kokum fruit rind. J. Food Sci. Technol. 1982,<br />

19, 97-100.<br />

(44) Singh, R. P.; Jayaprakasha, G. K.; Sakariah, K. K. (-)-<br />

Hydroxycitric acid from Garcinia cambogia. Biol. Memoirs<br />

1995, 21, 27-33.<br />

(45) Balasubramanyam, K.; Ch<strong>and</strong>rasekhar, B.; Ramadoss, C. S.; Rao,<br />

P. V. S. Soluble double metal salt <strong>of</strong> group IA <strong>and</strong> IIA <strong>of</strong> (-)-<br />

hydroxycitric acid, process <strong>of</strong> preparing the same <strong>and</strong> its use in<br />

beverages <strong>and</strong> other food products without effecting their flavor<br />

<strong>and</strong> properties. U.S. Patent 6160172, 2000.<br />

(46) Ibnusaud, I.; Puthiaparampil, T. T.; Thomas, B. Convenient<br />

method for the large-scale isolation <strong>of</strong> Garcinia acid. U.S. Patent<br />

6147228, 2000.<br />

(47) Watson, J. A.; Fang, M.; Lowenstein, J. M. Tricarballylate <strong>and</strong><br />

hydroxycitrate: Substrate <strong>and</strong> inhibition <strong>of</strong> ATP:citrate oxaloacetatae<br />

lyase. Arch. Biochem. Biophys. 1969, 135, 209-217.<br />

(48) Lowenstein, J. M. Experiments with (-)-hydroxycitrate. In<br />

Essays in Cell Metabolism; Burtley, W., Kornberg, H. L., Quayle,<br />

J. R., Eds.; Wiley-Interscience: New York, 1970; pp 153-166.<br />

(49) Cheema-Dhadli, S.; Halperin, M. L.; Lezn<strong>of</strong>f, C. C. Inhibition<br />

<strong>of</strong> enzymes which interact with citrate by (-)-hydroxycitrate<br />

<strong>and</strong> 1,2,3-tricarboxybenzene. Eur. J. Biochem. 1973, 38, 98-<br />

102.<br />

(50) Szutowicz, A.; Stepien, M.; Lysiak, W.; Angielski, S. Effect <strong>of</strong><br />

(-)-hydroxycitrate on the activities <strong>of</strong> ATP citrate lyase <strong>and</strong> the<br />

enzymes <strong>of</strong> acetyl-CoA metabolism in rat brain. Acta Biochim<br />

Pol. 1976, 23, 227-234.<br />

(51) Sullivan, A. C.; Singh, M.; Sere, P. A.; Glusker, J. P. Reactivity<br />

<strong>and</strong> inhibitor potential <strong>of</strong> hydroxycitrate isomers with citrate<br />

synthase, citrate lyases <strong>and</strong> ATP citrate lyases. J. Biol. Chem.<br />

1977, 252, 7583-7590.<br />

(52) H<strong>of</strong>fman, G. E.; Andres, H.; Weiss, L.; Kreisel, C.; S<strong>and</strong>er, R.<br />

Properties <strong>and</strong> organ distribution <strong>of</strong> ATP citrate (pro-3S)-lyase.<br />

Biochim Biophys. Acta 1980, 620, 151-158.<br />

(53) Watson, J. A.; Lowenstein, J. M. Citrate <strong>and</strong> the conversion <strong>of</strong><br />

carbohydrate into fat. J. Biol. Chem. 1970, 245, 5993-6002.<br />

(54) Lowenstin, J. M. Citrate <strong>and</strong> the conversion <strong>of</strong> carbohydrate into<br />

fat. Biochem. Soc. Symp. 1968, 27, 61-86.<br />

(55) Lehninger, A. L. <strong>Biochemistry</strong>, 2nd ed.; Kalyani Publishers: New<br />

Delhi, India, 1983; pp 659-691.<br />

(56) Stryer, L. <strong>Biochemistry</strong>, 3rd ed.; Freeman: New York, 1988;<br />

pp 487-488.<br />

(57) Rawn, J. D. <strong>Biochemistry</strong>; Neil Patterson: Burlington, NC, 1997;<br />

pp 421-455.<br />

(58) Srere, P. A. The citrate cleavage enzyme. I. Distribution <strong>and</strong><br />

purification. J. Biol. Chem. 1959, 234, 2544-2547.<br />

(59) Spencer, A.; Corman, L.; Lowenstein, J. M. Citrate <strong>and</strong> the<br />

conversion <strong>of</strong> carbohydrate into fat. Biochem. J. 1964, 93, 378-<br />

388.<br />

(60) D’Adamo, A. F., Jr.; Haft, D. E. An alternate <strong>of</strong> R-ketoglutarate<br />

catabolism in the isolated, perfused rat liver. J. Biol. Chem. 1965,<br />

240, 613-617.<br />

(61) Kornacker, M. S.; Lowenstein, J. M. Citrate <strong>and</strong> the conversion<br />

<strong>of</strong> carbohydrate into fat. Biochem. J. 1965, 94, 209-215.<br />

(62) Bressler, R.; Brendel, K. The role <strong>of</strong> carnitine <strong>and</strong> carnitine<br />

acyltransferse in biological acetylations <strong>and</strong> fatty acid synthesis.<br />

J. Biol. Chem. 1966, 241, 4092-4097.<br />

(63) Linn, T. C.; Srere, P. A. Identification <strong>of</strong> ATP citrate lyase as a<br />

phosphoprotein. J. Biol. Chem. 1979, 254, 1691-1698.<br />

(64) Daikuhara, Y.; Tsunemi, T.; Takeda, Y. The role <strong>of</strong> ATP citrate<br />

lyase in the transfer <strong>of</strong> acetyl groups in rat liver. Biochim.<br />

Biophys. Acta 1968, 158, 51-61.<br />

(65) Sullivan, A. C.; Hamilton, J. G.; Miller, O. N.; Wheatley, V. R.<br />

Inhibition <strong>of</strong> lipogenesis in rat liver by (-)-hydroxycitrate. Arch.<br />

Biochem. Biophys. 1972, 150, 183-190.<br />

(66) Sullivan, A. C.; Triscari, J.; Hamilton, J. G.; Miller, O. N.;<br />

Wheatley, V. R. Effect <strong>of</strong> (-)-hydroxycitrate upon the accumulation<br />

<strong>of</strong> lipid in rat. I. Lipogenesis. Lipids 1974, 9, 121-<br />

128.


20 J. Agric. Food Chem., Vol. 50, No. 1, 2002 Reviews<br />

(67) Sullivan, A. C.; Triscari, J.; Hamilton, J. G.; Miller, O. N. Effect<br />

<strong>of</strong> (-)-hydroxycitrate upon the accumulation <strong>of</strong> lipid in the rat.<br />

II. Appetite. Lipids 1974, 9, 129-134.<br />

(68) Sullivan, A. C. Effect <strong>of</strong> (-)-hydroxycitrate on lipid metabolism.<br />

In Modification <strong>of</strong> Lipid Metabolism; Perks, E. G., Witting, L.,<br />

Eds.; Academic Press: New York, 1984; pp 143-174.<br />

(69) Beynen, A. C.; Geelen, M. J. Effect <strong>of</strong> insulin <strong>and</strong> glucagon on<br />

fatty acid synthesis from acetate by hepatocytes incubated with<br />

(-)-hydroxycitrate. Endokrinologie 1982, 79, 308-310.<br />

(70) Barth, C.; Hackenschmidt, J.; Ullmann, H.; Decker, K. Inhibition<br />

<strong>of</strong> cholesterol synthesis by (-)-hydroxycitrate in perfused rat<br />

liver. Evidence for an extramitochondrial mevalonate synthesis<br />

from acetyl coenzyme A. FEBS Lett. 1972, 22, 343-346.<br />

(71) Gregolin, C.; Ryder, E.; Warner, R. C.; Kleinschmidt, A. K.;<br />

Chang, H. C.; Lane, D. M. Liver acetyl coenzyme A carboxylase.<br />

II. Further molecular characterization. J. Biol. Chem. 1968, 243,<br />

4236-4245.<br />

(72) Hashimoto, T.; Numa, S. Kinetic studies on the reaction<br />

mechanism <strong>and</strong> the citrate activation <strong>of</strong> liver acetyl coenzyme<br />

A carboxylase. Eur. J. Biochem. 1971, 18, 319-331.<br />

(73) Hackenschmidt, J.; Barth, C.; Decker, K. Stimulation <strong>of</strong> acetyl-<br />

CoA carboxylase by (-)-hydroxycitrate. FEBS Lett. 1972, 27,<br />

131-133.<br />

(74) Sugden, M. C.; Steare, S. E.; Watts, D. I.; Palmer, T. N.<br />

Interactions between insulin <strong>and</strong> thyroid hormone in the control<br />

<strong>of</strong> lipogenesis. Biochem. J. 1983, 210, 937-944.<br />

(75) Sugden, M. C.; Watts, D. I.; Marshall, C. E.; McCormack, J. G.<br />

Brown-adipose-tissue lipogenesis in starvation: Effects <strong>of</strong> insulin<br />

<strong>and</strong> (-)-hydroxycitrate. Biosci. Rep. 1982, 2, 289-297.<br />

(76) Smith, S. Lipogenesis in rabbit adipose tissue. J. Lipid Res. 1975,<br />

16, 324-331.<br />

(77) Sugden, M. C.; Watts, D. I.; West, P. S.; Palmer, T. N. Proline<br />

<strong>and</strong> hepatic lipogenesis. Biochim. Biophys. Acta 1984, 798, 368-<br />

373.<br />

(78) Palmer, T. N.; Caldecourt, M. A.; Watts, D. I.; Sugden, M. C.<br />

Inhibition <strong>of</strong> lipogenesis by vassopressin <strong>and</strong> angiotensin II, in<br />

glycogen-depleted hepatocytes. Biosci. Rep. 1983, 3, 1063-1070.<br />

(79) Evans, R. M.; Scholz, R. W. Pulmonary fatty acid synthesis. I.<br />

Mitochondrial acetyl transfer by rat lung in Vitro. Am. J. Physiol.<br />

1977, 232, E358-363.<br />

(80) Todhunter, D. A.; Scholz, R. W. In ViVo incorporation <strong>of</strong> tritium<br />

from 3 H 2O into pulmonary lipids <strong>of</strong> meal-fed <strong>and</strong> starved rats.<br />

Am. J. Physiol. 1996, 239, E407-411.<br />

(81) Sheehan, P. M.; Yeh, Y. Y. Pathways <strong>of</strong> acetyl-CoA production<br />

for lipogenesis from acetoacetate, beta-hydroxybutyrate, pyruvate<br />

<strong>and</strong> glucose in neonatal rat lung. Lipids 1984, 19, 103-108.<br />

(82) Crabtree, B.; Souter, M. J.; Anderson, S. E. Evidence that the<br />

production <strong>of</strong> acetate in rat hepatocytes is a predominantly<br />

cytoplasmic process. Biochem. J. 1989, 257, 673-678.<br />

(83) Groot, P. H. E.; Scholte, H. R.; Hulsmann, W. C. Fatty acid<br />

activation: Specificity, localization <strong>and</strong> function. AdV. Lipid Res.<br />

1976, 14, 75-126.<br />

(84) Vina, J. R.; Williamson, D. H. Effects <strong>of</strong> lactation on L-leucine<br />

metabolism in the rat. Biochem. J. 1981, 194, 941-947.<br />

(85) Mathias, M. M.; Sullivan, A. C.; Hamilton, J. G. Fatty acid <strong>and</strong><br />

cholesterol synthesis from specifically labeled leucine by isolated<br />

rat hepatocytes. Lipids 1981, 16, 739-743.<br />

(86) Hood, R. L.; Beitz, D. C.; Johnson, D. C. Inhibition by potential<br />

metabolic inhibitors <strong>of</strong> in Vitro adipose tissue lipogenesis. Comp.<br />

Biochem. Physiol. 1985, 81B, 667-670.<br />

(87) Hagve, T. A.; Christopherson, B. O. Linolenic acid desaturation<br />

<strong>and</strong> chain elongation <strong>and</strong> rapid turnover <strong>of</strong> phospholipid n-3-<br />

fatty acids in isolated rat liver cells. Biochim. Biophys. Acta 1983,<br />

753, 339-349.<br />

(88) Vicario, C.; Medina, M. Metabolism <strong>of</strong> lactate in the rat brain<br />

during the early neonatal period. J. Neurochem. 1992, 59, 32-<br />

40.<br />

(89) Patel, M. S.; Owen, O. E. Lipogenesis from ketone bodies in<br />

rat brain. Evidence for the conversion <strong>of</strong> acetoacetate into acetylcoenzyme<br />

A in the cytosol. Biochem. J. 1976, 156, 603-607.<br />

(90) Sterling, G. H.; O’Neilil, J. J. Citrate as the precursor <strong>of</strong> the<br />

acetyl moiety <strong>of</strong> acetylcholine. J. Neurochem. 1978, 31, 525-<br />

530.<br />

(91) Wheatley, V. R.; Brind, J. L. Sebaceous gl<strong>and</strong> differentiation,<br />

III. The uses <strong>and</strong> limitations <strong>of</strong> freshly isolated mouse preputial<br />

gl<strong>and</strong> cells for the in Vitro study <strong>of</strong> hormone <strong>and</strong> drug action. J.<br />

InVest. Dermatol. 1981, 76, 293-296.<br />

(92) Hamilton, J. G.; Sullivan, A.C.; Kritchevsky, D. Hypolipidemic<br />

activity <strong>of</strong> (-)-hydroxycitrate. Lipids 1977, 12, 1-9.<br />

(93) Pullinger, C. R.; Gibbons, G. F. The role <strong>of</strong> substrate supply in<br />

the regulation <strong>of</strong> cholesterol biosynthesis in rat hepatocytes.<br />

Biochem. J. 1983, 210, 625-632.<br />

(94) Brunengraber, H.; Sabine, J. R.; Boutry, M.; Lowenstein, J. M.<br />

3-Hydroxysterol synthesis by liver. Arch. Biochem. Biophys.<br />

1972, 150, 392-396.<br />

(95) Brunengraber, H.; Boutry, M.; Lowenstein, J. M. Fatty acid, 3-βhydroxysterol<br />

<strong>and</strong> ketone synthesis in the perfused rat liver. Eur.<br />

J. Biochem. 1978, 82, 373-384.<br />

(96) Patterson, C. E.; Davis, K. S.; Rhodes, R. A. Regulation <strong>of</strong> fetlal<br />

lung disaturated phosphatidylcholine synthesis by de novo<br />

palmitate supply. Biochim. Biophys. Acta 1988, 958, 60-69.<br />

(97) Berkhout, J. A.; Havekes, L. M.; Pearce, N. J.; Groot, P. H. The<br />

effect <strong>of</strong> (-)-hydroxycitrate on the activity <strong>of</strong> the low-density<br />

lipoprotein receptor <strong>and</strong> 3-hydroxy-3-methyl-glutaryl-CoA reductase<br />

levels in the human hepatoma cell line HepG2. Biochem.<br />

J. 1990, 272, 181-186.<br />

(98) Hildebr<strong>and</strong>t, L. A.; Spennetta, T.; Elson, C.; Shrago, E. Utilization<br />

<strong>and</strong> preferred metabolic pathway <strong>of</strong> ketonebodies for lipid<br />

synthesis by isolated rat hepatoma cells. Am. J. Physiol. 1995,<br />

269, C22-27.<br />

(99) Lligona-Trulla, L.; Arduini, A.; Aldaghlas, A. T.; Calvani, M.;<br />

Kelleher, J. K. Acetyl-L-carnitine flux to lipids in cells estimated<br />

using isotopomer spectral analysis. J. Lipid Res. 1997, 38, 1454-<br />

1462.<br />

(100) Noboru, H. Inhibition <strong>of</strong> lipogenesis <strong>and</strong> stimulation <strong>of</strong> lipolysis<br />

in 3T3L1 cells by Garcinia extract. Nippon Kasei Gakkaishi<br />

1998, 49, 889-892.<br />

(101) Lowenstein, J. M. Effect <strong>of</strong> (-)-hydroxycitrate on fatty acid<br />

synthesis by rat liver in ViVo. J. Biol. Chem. 1971, 246, 629-<br />

632.<br />

(102) Triscari, J.; Sullivan, A. C. Comparative effects <strong>of</strong> (-)-<br />

hydroxycitrate <strong>and</strong> (+)-allo-hydroxycitate on acetyl CoA carboxylase<br />

<strong>and</strong> fatty acid <strong>and</strong> cholesterol synthesis in ViVo. Lipids<br />

1977, 12, 357-363.<br />

(103) Saha, A. K.; Vavvas, D.; Kurowshi, T. G.; Apanidis, A.; Witters,<br />

L. A.; Shafrir, E.; Ruderman, N. B. Malonyl-CoA regulation in<br />

skeletal muscles, its link to cell citrate <strong>and</strong> the glucose-fatty acid<br />

cycle. Am. J. Physiol. 1997, 272, E641-648.<br />

(104) Sullivan, A. C.; Triscari, J. Metabolic regulation as a control<br />

for lipid disorders, I. Influence <strong>of</strong> (-)-hydroxycitrate on experimentally<br />

induced obesity in the rodent. Am. J. Clin. Nutr. 1977,<br />

30, 767-776.<br />

(105) Chee, H.; Romos, D. R.; Leveille, G. A. Influence <strong>of</strong> (-)-<br />

hydroxycitrate on lipogenesis in chickens <strong>and</strong> rats. J. Nutr. 1977,<br />

107, 112-119.<br />

(106) Sullivan, A. C.; Triscari, J.; Spiegel, J. E. Metabolic regulation<br />

as a control for lipid disorders. II. Influence <strong>of</strong> (-)-hydroxycitrate<br />

on genetically <strong>and</strong> experimentally induced hypertriglyceridemia<br />

in the rat. Am. J. Clin. Nutr. 1977, 30, 777-784.<br />

(107) Greenwood, M. R.; Cleary, M. P.; Gruen, R.; Blasé, D.; Stern,<br />

J. S.; Triscari, J.; Sullivan, A. C. Effect <strong>of</strong> (-)-hydroxycitrate<br />

on development <strong>of</strong> obesity in the Zucker obese rat. Am. J.<br />

Physiol. 1981, 240, E72-78.<br />

(108) Rao, R. N.; Sakariah, K. K. Lipid-lowering <strong>and</strong> antiobesity effect<br />

<strong>of</strong> (-)-hydroxycitric acid. Nutr. Res. 1988, 8, 209-212.<br />

(109) Brunengraber, H.; Lowenstein, J. M. Effect <strong>of</strong> (-)-hydroxycitrate<br />

on ketone production by the perfused liver. FEBS Lett. 1976,<br />

65, 251-253.<br />

(110) Leighton, B.; Nicholas, A. R.; Pogson, C. I. The pathway <strong>of</strong><br />

ketogenesis in rumen epithelium <strong>of</strong> the sheep. Biochem. J. 1983,<br />

216, 769-772.


Reviews J. Agric. Food Chem., Vol. 50, No. 1, 2002 21<br />

(111) Xu, Z. X.; Smart, D. A.; Rooney, S. A. Glucocorticoid induction<br />

<strong>of</strong> fatty acid synthase mediates the stimulatory effect <strong>of</strong> the<br />

hormone on cholinephosphate cytidylyltransferase activity on<br />

fetal rat lung. Biochim. Biophys. Acta 1990, 1044, 70-76.<br />

(112) Garl<strong>and</strong>, P. B.; R<strong>and</strong>le, P. J.; Newsholme, E. A. Citrate as an<br />

intermediary in the inhibition <strong>of</strong> phosfructokinase in rat heart<br />

muscle by fatty acids, ketone bodies, pyruvate in diabetes <strong>and</strong><br />

starvation. Nature 1963, 200, 169-170.<br />

(113) Paramaggiani, A.; Bowman, R. H. Regulation <strong>of</strong> phosph<strong>of</strong>ructokinase<br />

activity by citrate in normal <strong>and</strong> diabetic muscle.<br />

Biochem. Biophys. Res. Commun. 1963, 12, 268-273.<br />

(114) Zammit, V. A. Effects <strong>of</strong> citrate on phosph<strong>of</strong>ructokinase from<br />

lactating rat mammary gl<strong>and</strong> acini. FEBS Lett. 1979, 108, 193-<br />

196.<br />

(115) Bloxham, D. P.; Lardy, H. A. In The Enzymes, 3rd ed.; Boyer,<br />

P. D., Ed.; Academic Press: New York, 1995; Vol. 8, pp 239-<br />

278.<br />

(116) McCune, S. A.; Foe, L. G.; Kemp, R. G.; Jurin, R. R.<br />

Aurintricarboxylic acid is a potent inhibitor <strong>of</strong> phosph<strong>of</strong>ructokinase.<br />

Biochem. J. 1989, 259, 925-927.<br />

(117) Melnick, J. Z.; Srere, P. A.; Elshourbagy, N. A.; Moe, O. W.;<br />

Preisig, P. A.; Alpern, R. J. Adenosine triphosphate citrate lyase<br />

mediates hypocitraturia in rats. J. Clin. InVest. 1996, 98, 2381-<br />

2387.<br />

(118) Board, M.; Newsholme, E. Hydroxycitrate causes altered pyruvate<br />

metabolism by tumourigenic cells. Biochem. Mol. Biol. Int.<br />

1996, 40, 1047-1056.<br />

(119) Panksepp, J.; Pollack, A.; Meeker, R. B.; Sullivan, A. C. (-)-<br />

Hydroxycitrate <strong>and</strong> conditioned aversions. Pharmacol. Biochem.<br />

BehaV. 1977, 6, 683-687.<br />

(120) Van-Itallie, T. B.; Beaudion, R.; Mayer, J. Arteriovenous glucose<br />

differences metabolic hypoglycemia <strong>and</strong> food intake in man. J.<br />

Clin. Nutr. 1953, 1, 208-217.<br />

(121) Davis, J. D.; Gallaghar, R. L.; Ladove, R. Food intake controlled<br />

by a blood factor. Science 1967, 156, 1247-1248.<br />

(122) Schalley, A. W.; Redding, T. W.; Lucien, H. W.; Meyer, J.<br />

Enterogastrone inhibits eating by fasted rats. Science 1967, 157,<br />

210-211.<br />

(123) Fernstrom, J. D.; Wurtman, R. J. Brain serotonin content:<br />

Physiological dependence on plasma tryptophan levels. Science<br />

1971, 173, 149-152.<br />

(124) Fernstrom, J. D.; Wurtman, R. J. Brain serotonin content:<br />

Increase following ingestion <strong>of</strong> carbohydrate diet. Science 1971,<br />

174, 1023-1025.<br />

(125) Fernstrom, J. D.; Wurtman, R. J. Elevation <strong>of</strong> plasma tryptophan<br />

by insulin in rat. Metabolism 1972, 21, 337-342.<br />

(126) Fernstrom, J. D.; Wurtman, R. J. Brain serotonin content:<br />

Physiological regulation by plasma neutral amino acids. Science<br />

1972, 178, 414-416.<br />

(127) Garattini, S.; Mennini, T.; Bendotti, C.; Invernizzi, R.; Samanin,<br />

R. Neurochemical mechanism <strong>of</strong> action <strong>of</strong> drugs which modify<br />

feeding via the serotoninergic system. Appetite 1988, 7 (Suppl.),<br />

15-38.<br />

(128) Zigmond, M. J.; Stricker, E. M. Deficits in feeding behavior<br />

after intravascular injection <strong>of</strong> 6-hydroxydopamine in rats.<br />

Science 1972, 177, 1211-1214.<br />

(129) Ricny, J.; Tucek, S. Relation between the content <strong>of</strong> acetylcoenzyme<br />

A <strong>and</strong> acetyl choline in brain slices. Biochem. J. 1980,<br />

188, 683-688.<br />

(130) Endemann, G.; Brunengraber, H. The source <strong>of</strong> acetyl coenzyme<br />

A for acetylcholine synthesis in the perfused rat phrenic nerves<br />

hemidiaphragm. J. Biol. Chem. 1980, 255, 11091-11093.<br />

(131) Gibson, G. E.; Shimada, M. Studies on metabolic pathway <strong>of</strong><br />

the acetyl group for acetylcholine synthesis. Biochem. Pharmacol.<br />

1980, 29, 167-174.<br />

(132) Tucek, S.; Dolezal, V.; Sullivan, A. C. Inhibition <strong>of</strong> synthesis<br />

<strong>of</strong> acetylcholine in rat brain slices by (-)-hydroxycitrate <strong>and</strong><br />

citrate. J. Neurochem. 1981, 36, 1331-1337.<br />

(133) Sterling, G. H.; McCafferty, M. R.; O’Neill, J. J. β-Hydroxybutyrate<br />

as a precursor to the acetyl moiety <strong>of</strong> acetylcholine. J.<br />

Neurochem. 1981, 37, 1250-1259.<br />

(134) Ricny, J.; Tucek, S. Acetyl coenzyme A <strong>and</strong> acetylcholine in<br />

slices <strong>of</strong> rat caudate nucleie incubated with (-)-hydroxycitrate,<br />

citrate <strong>and</strong> EGTA. J. Neurochem. 1982, 39, 668-673.<br />

(135) Gibson, G. E.; Peterson, C. Acetylcholine <strong>and</strong> oxidative metabolism<br />

in septum <strong>and</strong> hippocampus, in Vitro. J. Biol. Chem.<br />

1983, 258, 1142-1145.<br />

(136) Szutowicz, A.; Bielarczyk, H.; Sklilimowska, H. Effect <strong>of</strong><br />

dichloroacetate on acetyl-CoA content <strong>and</strong> acetylcholine synthesis<br />

in rat brain synaptosomes. Neurochem. Res. 1994, 19,<br />

1107-1112.<br />

(137) Burton, M. D.; Nouri, M.; Kazemi, H. Acetylcholine <strong>and</strong> central<br />

respiratory control: perturbations <strong>of</strong> acetylcholine synthesis in<br />

the isolated brainstem <strong>of</strong> the neonatal rat. Brain Res. 1995, 670,<br />

39-47.<br />

(138) McGarry, J. D.; Leatherman, G. F.; Foster, D. W. Carnitine<br />

palmitoyl transferase I. The site <strong>of</strong> inhibition <strong>of</strong> hepatic fatty<br />

acid oxidation by malonyl-CoA. J. Biol. Chem. 1978, 253, 4128-<br />

4136.<br />

(139) McGarry, J. D.; Foster, D. W. In support <strong>of</strong> the roles <strong>of</strong> malonyl-<br />

CoA <strong>and</strong> carnitine acyltransferase I in the regulation <strong>of</strong> hepatic<br />

fatty acid oxidation <strong>and</strong> ketogenesis. J. Biol. Chem. 1979, 254,<br />

8163-8168.<br />

(140) Ruderman, N. B.; Saha, A. K.; Vavvas, D.; Witters, L. A.<br />

Malonyl-CoA fuel sensing <strong>and</strong> insulin resistance. Am. J. Physiol.<br />

1999, 276, E1-E18.<br />

(141) Yang, X.; Kow, L. M.; Funabashi, T.; Mobbs, C. V. Hypothalamic<br />

glucose sensor: Similarities to <strong>and</strong> differences from<br />

pancreatic mechanisms. Diabetes 1999, 48, 1763-1772.<br />

(142) Chen, S.; Ogawa, A.; Ohneda, M.; Unger, R. H.; Foster, D. W.;<br />

McGarry, J. D. More direct evidence for a malonyl-CoA carnitine<br />

palmitoyltransferase I interaction as a key event in pancreatic<br />

cell signaling. Diabetes 1994, 43, 878-883.<br />

(143) Sullivan, A. C.; Triscari, J.; Miller, O. N. The influence <strong>of</strong> (-)-<br />

hydroxycitrate on in ViVo rates <strong>of</strong> hepatic glycogenesis <strong>and</strong><br />

cholesterogenesis. Fed. Proc. 1974, 33, 656.<br />

(144) Sullivan, A. C.; Triscari, J.; Cheng, L. Appetite regulation by<br />

drugs <strong>and</strong> endogenous substances. In Nutrition <strong>and</strong> Drugs<br />

(Current Concepts in Nutrition, Vol. 12); Winick, M., Ed.;<br />

Wiley: New York, 1983; pp 139-167.<br />

(145) Sullivan, A. C.; Gruen, R. K. Mechanisms <strong>of</strong> appetite modulation<br />

by drugs. Fed. Proc. 1985, 44, 139-144.<br />

(146) Hellerstein, M. K.; Xie, Y. The indirect pathway <strong>of</strong> hepatic<br />

glycogen synthesis <strong>and</strong> reduction <strong>of</strong> food intake by metabolic<br />

inhibitors. Life Sci. 1993, 53, 1833-1845.<br />

(147) McCarty, M. F. Promotion <strong>of</strong> hepatic lipid oxidation <strong>and</strong><br />

gluconeogenesis a strategy for appetite control. Med. Hypotheses<br />

1994, 42, 215-225.<br />

(148) McCarty, M. F.; Gustin, J. C. Pyruvate <strong>and</strong> hydroxycitrate/<br />

carnitine may synergise to promote reverse electron transport in<br />

hepatocyte mitochondria, effectively uncoupling the oxidation<br />

<strong>of</strong> fatty acids. Med. Hypotheses 1999, 52, 407-416.<br />

(149) McCarty, M. F. Reduction <strong>of</strong> free fatty acids may ameliorate<br />

risk factors associated with abdominal obesity. Med. Hypotheses<br />

1995, 44, 278-286.<br />

(150) McCarty, M. F. Inhibition <strong>of</strong> citrate lyase may aid aerobic<br />

endurance. Med. Hypotheses 1995, 45, 247-254.<br />

(151) McCarty, M. F. Optimizing exercise for fat loss. Med. Hypotheses<br />

1995, 44, 325-330.<br />

(152) Kriketos, A. D.; Thompson, H. R.; Greene, H.; Hill, J. O. (-)-<br />

Hydroxycitric acid does not affect energy expenditure <strong>and</strong><br />

substrate oxidation in adult males in a post-absorptive state. Int.<br />

Obes. Relat. Metab. Disord. 1999, 23, 867-873.<br />

(153) Ishihara, K.; Oyaizu, S.; Onuki, K.; Lim, K.; Fushik, T. Chronic<br />

(-)-hydroxycitrate administration spares carbohydrate utilization<br />

<strong>and</strong> promotes lipid oxidation during exercise in mice. J. Nutr.<br />

2000, 130, 2990-2995.<br />

(154) Clouatre, D.; Rosenbaum, M. The Diet <strong>and</strong> Health Benefits <strong>of</strong><br />

<strong>HCA</strong> (Hydroxycitric Acid); Keats Publishing: New Cannan, CT,<br />

1994.


22 J. Agric. Food Chem., Vol. 50, No. 1, 2002 Reviews<br />

(155) Heymsfield, S. B.; Allison, D. B.; Vasselli, J. R.; Pietobelli, D.<br />

G.; Nunthe, C. Garcinia cambogia (Hydroxycitric acid) as a<br />

potential antiobesity agent. A r<strong>and</strong>omized control trial. JAMA-<br />

J. Am. Med. Assoc. 1998, 280, 1596-1600.<br />

(156) Badmaev, V.; Majeed, M. Open field, physician controlled,<br />

clinical evaluation <strong>of</strong> botanical weight loss formula Citrin.<br />

Presented at Nutracon; Nutraceuticals, Dietary Supplements <strong>and</strong><br />

Functional Foods, Las Vegas, NV, July 11-13, 1995.<br />

(157) Conte, A. A. A non-prescription alternative on weight reduction<br />

therapy. Am. Barariatr. Med. Summer 1993, 17-19.<br />

(158) Thom, E. Hydroxycitrate (<strong>HCA</strong>) in the treatment <strong>of</strong> obesity. Int.<br />

J. Obes. 1996, 20 (Suppl. 4), 48.<br />

(159) Rothacker, D. Q.; Waitman, B. E. Effectiveness <strong>of</strong> a Garcinia<br />

cambogia <strong>and</strong> natural caffeine <strong>com</strong>bination in weight loss: A<br />

double-blind placebo-controlled pilot study. Int. J. Obes. 1997,<br />

21 (Suppl. 2), 53.<br />

(160) Girola, M.; De Bernardi, M.; Contos, S. Dose effect on lipid<br />

lowering activity <strong>of</strong> a dietary intetrator, (chitosan, Garcinia<br />

cambogia extract <strong>and</strong> chrome). Acta Toxicol. Ther. 1996, 17,<br />

25-40.<br />

(161) Hobbs, L. S. (-)-Hydroxycitrate (<strong>HCA</strong>). In The New Diet Pills;<br />

Pragmatic Press: Irvine, CA, 1994; pp 161-174.<br />

(162) Ramos, R. R.; Saenz, F. J.; Alarcon, A. Extract <strong>of</strong> Garcinia<br />

cambogia in the control <strong>of</strong> obesity (in Spanish). InVest. Med.<br />

Int. 1996, 22, 97-100.<br />

(163) Vasselli, J. R.; Shane, E.; Boozer, C. N.; Heymsfield, S. B.<br />

Garcinia cambogia extract exhibits body weight gain via<br />

increased energy expenditure (EE) in rats. FASEB J. 1998, 12<br />

(Part I), A 505.<br />

(164) Puttaparthi, K. R.; T. Elshourbagy, N. A.; Levi, M. Renal ATP<br />

citrate lyase (ATP CL) protein localizes throughout the nephron<br />

<strong>and</strong> increases only in the proximal tube with chronic metabolic<br />

acidosis (CMA). Presented at the Pediatric Academic Societies,<br />

Annual Meeting, May 2, San Francisco, CA, 1999; Abstract.<br />

(165) Sawada, H.; Tomi, H.; Tamura, K. Effects <strong>of</strong> liquid Garcinia<br />

extract <strong>and</strong> soluble Garcinia powder on body weight change: a<br />

possible material for suppressing fat accumulation. Nihon<br />

Yukagaku Kaishi 1997, 46, 1467-1474.<br />

(166) McCarty, M. F.; Majeed, M. The pharmacology <strong>of</strong> citrin. In<br />

Citrin: A ReVolutionary Herbal Approach to Weight Management;<br />

Majeed, M., Ed.; New Editions Publishing: Burlingame,<br />

CA, 1994; pp 34-51.<br />

(167) McCarty, M. F. Utility <strong>of</strong> metformin as adjunct to hydroxycitrate/<br />

carnitine for reducing body fat in diabetics. Med. Hypotheses<br />

1998, 51, 399-403.<br />

(168) Prentki, M.; Corkey, B. K. Are beta-cell signaling molecules<br />

malonyl-CoA <strong>and</strong> cytosolic long chain acetyl CoA implicated<br />

in multiple tissue defects <strong>of</strong> obesity <strong>and</strong> NIDDM Diabetes 1996,<br />

45, 273-283.<br />

(169) Brun, T.; Roche, E.; Assimacopoulos-Jeannet, F.; Corkey, B.<br />

E.; Kim, K. H.; Prentki, M. Evidence for anaplerotic/malonyl<br />

CoA pathway in pancreatic beta cell nutrient signaling. Diabetes<br />

1996, 45, 190-198.<br />

(170) Corkey, B. E.; Glennon, M. C.; Chen, K. S.; Deeney, J. T.;<br />

Matschinsky, F. M.; Prentki, M. A role for malonyl-CoA in<br />

glucose-stimulated insulin secretion from clonal pancreatic beta<br />

cells. J. Biol. Chem. 1989, 264, 21608-21612.<br />

(171) Sener, A.; Malaisse, W. J. Hexose metabolism on pancreatic<br />

islets. Effect <strong>of</strong> (-)-hydroxycitrate upon fatty acid synthesis <strong>and</strong><br />

insulin release on glucose stimulated islets. Biochimie 1991, 73,<br />

1287-1290.<br />

(172) Prentki, M.; Vischer, S.; Glenon, M. C.; Regazz, R.; Deeney, J.<br />

T.; Corkey, B. E. Malonyl-CoA <strong>and</strong> long chain acyl-CoA esters<br />

as metabolic coupling factors in nutrient-induced insulin secretion.<br />

J. Biol. Chem. 1992, 267, 5802-5810.<br />

(173) Duan, C.; Winder, W. W. Control <strong>of</strong> malonyl-CoA by glucose<br />

<strong>and</strong> insulin in perfused skeletal muscle. J. Appl. Physiol. 1993,<br />

74, 2543-2547.<br />

(174) Saha, A. K.; Kurowski, T. G.; Ruderman, N. B. A malonyl CoA<br />

fuel sensing mechanism in muscle: Effects <strong>of</strong> insulin glucose<br />

<strong>and</strong> denervation. Am. J. Physiol. 1995, 269, E283-289.<br />

(175) Prentki, M. Tornhein, K.; Corkey, B. E. Signal transudation<br />

mechanisms in nutrient induced insulin secretion. Diabetologia<br />

1997, 40 (Suppl. 2), S32-41.<br />

(176) Zhang, S.; Kim, K. H. Essential role <strong>of</strong> acetyl-CoA carboxylase<br />

in the glucose induced insulin secretion in a pancreatic beta cell<br />

line. Cell Signal 1998, 10, 35-42.<br />

(177) Ruderman, N. B.; Saha, A. K.; Vavvas, D.; Kurowski, T.;<br />

Laybutt, D. R.; Schmitz-Peiffer, C.; Biden, T.; Kraegen, E. N.<br />

Malonyl-CoA as a metabolic switch <strong>and</strong> a regulator <strong>of</strong> insulin<br />

sensitivity. AdV. Exp. Biol. 1998, 441, 263-270.<br />

Received for review June 8, 2001. Revised manuscript received<br />

September 25, 2001. Accepted September 26, 2001.<br />

JF010753K

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!